US20090076250A1 - Substance that Specifically Recognizes PD-1 - Google Patents

Substance that Specifically Recognizes PD-1 Download PDF

Info

Publication number
US20090076250A1
US20090076250A1 US12/057,637 US5763708A US2009076250A1 US 20090076250 A1 US20090076250 A1 US 20090076250A1 US 5763708 A US5763708 A US 5763708A US 2009076250 A1 US2009076250 A1 US 2009076250A1
Authority
US
United States
Prior art keywords
antibody
cells
substance
antibodies
manufactured
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US12/057,637
Other versions
US7858746B2 (en
Inventor
Tasuku Honjo
Shiro Shibayama
Masayoshi Matsuo
Takao Yoshida
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ono Pharmaceutical Co Ltd
Original Assignee
Ono Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ono Pharmaceutical Co Ltd filed Critical Ono Pharmaceutical Co Ltd
Priority to US12/057,637 priority Critical patent/US7858746B2/en
Publication of US20090076250A1 publication Critical patent/US20090076250A1/en
Application granted granted Critical
Publication of US7858746B2 publication Critical patent/US7858746B2/en
Adjusted expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/02Local antiseptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/626Diabody or triabody
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention is related to a substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker.
  • the immune system acquired the mechanism that can respond to various foreign antigens.
  • the mechanism brings about the diversity of an antigen receptor by recombination of V, (D) and J fragment in T cells and B cells. Although this mechanism brought a result that produces self-reactive lymphocytes simultaneously, these self-reactive lymphocytes are removed by the negative selection in the thymus or bone marrow, and are further controlled by the self-tolerance mechanism of clone removal or anergy in the periphery.
  • PD-1 is type I membrane protein of 55 kD belonging to an immunoglobulin family. Bothmouse PD-1 and human PD-1 consist of 288 amino acids, and have signal peptide at N terminal (20 amino acid) and hydrophobic region in the middle part, which is a transmembrane region (The EMBO J. 11 (11):3887-3895, 1992); Japanese patent Publication No. 5-336973; EMBL/GenBank/DDJB Acc. No. X67914, Genomics 23:704, 1994; Japanese patent Publication No. 7-291996 (U.S. Pat. No. 5,629,204).
  • PD-1 is expressed during a CD4 ⁇ CD8 ⁇ cell differentiation to a CD4+ CD8+ cell (Nishimura H. et. al., Int. Immunol. 8:773-780, 1996, Nishimura H. et. al., J. Exp. Med. 191:891-898, 2000).
  • PD-1 is expressed in T cells and B cells which were activated by the stimulus from an antigen receptor (Agata Y. et. al., Int. Immunol. 8:765-772, 1996), and in bone marrow cells including activated macrophage.
  • PD-1 has ITIM (Immunoreceptor tyrosine-based inhibitory motif) in its intracellular region, therefore it is considered to be a negative regulator in immune reaction.
  • PD-1 deficit mice develop a lupus-like autoimmune disease such as glomerular nephritis and arthritis (C57BL/6 genetic background) (Nishimura H. et. al., Int. Imuunol. 10:1563-1572, 1998, Nishimura H. et. al., Immunity 11:141-151, 1999) and dilated cardiomyopathy-like disease (BALB/c genetic background) (Nishimura H. et. al., Science 291:319-322, 2001), it became clear that PD-1 is a regulator of the development of autoimmune disease, especially one of self-tolerance in the periphery.
  • ITIM Immunoreceptor tyrosine-based inhibitory motif
  • PD-1 is the regulator of various autoimmune diseases, and that it is one of the genes that cause an autoimmune disease.
  • the medical treatment and diagnosis of suppression or enhancement of the immune function, infection, the rejection at the time of a transplant, a neoplasm, etc. could be performed, and as a result of repeating examination wholeheartedly, the inventors reached this invention concerning the substance which controls the function of PD-1.
  • TCR T cell receptor
  • BCR B cell receptor
  • PD-1 is controlling negatively various cells responsible for immunity, such as lymphocytes and myeloid cells etc.
  • ITIM Immunoreceptor tyrosine-based inhibitory motif
  • the inventors considered the molecular mechanism involved in the inhibitory signal transduction of PD-1 to be the recruit of de-phosphorylation enzymes (phosphatases). Therefore, it came to be considered by locating PD-1 near TCR or BCR that it can display the function of PD-1.
  • CD3 is the membrane protein expressed on a T cell, and is one component of the complexes that constitute TCR.
  • the divalent antibody was constructed by bridging anti-PD-1 antibody and anti-BCR or anti-CD3 antibody. In the present invention, this divalent antibody is called hybrid antibody. The inventors produced this hybrid antibody for the first time.
  • the present invention relates to,
  • a substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker, 2.
  • the substance according to (1) which is a divalent substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker, 3.
  • the substance according to (1) or (2) in which a membrane protein is a protein existing on a T cell membrane or B cell membrane, 4.
  • the substance according to (1) or (2) which comprises a substance that recognizes PD-1, a substance that recognizes a protein constituting T cell receptor complex or a substance that recognizes a protein constituting B cell receptor complex, and a linker, 5.
  • a linker comprises an organic compound, 9.
  • the substance according to any one of (1) to (5), in which a linker comprises a peptide, 10.
  • the substance according to any one of (1) to (5), in which a substance that recognizes PD-1 and in which a substance that recognizes a protein comprises two or more peptides including a heavy chain variable region and a light chain variable region of antibody, respectively, 12.
  • a pharmaceutical composition containing an effective dose of the substance according to (1) for the medical treatment and/or prevention of a disease in which PD-1 participates 14.
  • autoimmune disease is selected from the group consisting of glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis.
  • autoimmune disease is selected from the group consisting of glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact
  • a substance that recognizes PD-1 said by the present invention may be a substance which recognizes PD-1 specifically, for example, anti-PD-1 antibody, the fragment of anti-PD-1 antibody, PD-1 in itself, the fragment of PD-1, ligand of PD-1 (PD-L1 (Freeman G. J. et. al., J. Exp. Med. 192:1027-1034, 2000), PD-L2, PD-H3), the fragment thereof and a low molecule organic compound, etc.
  • PD-L1 Freeman G. J. et. al., J. Exp. Med. 192:1027-1034, 2000
  • PD-L2, PD-H3 ligand of PD-1
  • anti-PD-1 antibody produced by the hybridoma strain “J43”, which was deposited on May 30, 2001 in International Patent Organism Depositary, National Institute of Advanced Industrial Science and Technology, AIST Tsukuba Central 6, 1-1, Higashi 1-Chome Tsukuba-shi, Ibaraki-ken 305-8566 Japan as Accession number FERM P-18356, and transferred to international deposition on Jul. 16, 2002 as international deposition Accession number FERM BP-8118.
  • the Fab portion of the antibody it is not limited to this.
  • a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane said by the present invention may be a substance which recognizes a membrane protein specifically, for example, a substance which specifically recognizes a complex that constitute T cell receptor (TCR) (TCR complex) etc. expressed on T cells, or a substance which specifically recognizes a complex that constitute B cell receptor (BCR) (BCR complex) etc. expressed on B cells, such as the fragment of a protein constituting TCR complex, anti-TCR antibody, the fragment of anti-TCR antibody, the fragment of a protein constituting BCR complex, anti-BCR antibody and the fragment of anti-BCR antibody.
  • TCR T cell receptor
  • BCR BCR complex
  • the Fab portion of an anti-TCR antibody or the Fab portion of an anti-BCR antibody it is not limited to these.
  • anti-CD3 antibody ⁇ -CD3 ⁇ mAb, manufactured by Pharmingen
  • anti-IgG (H+L) polyclonal antibody manufactured by Zymed
  • a linker said by the present invention may be a substance which can connect the above mentioned substance recognizing PD-1 and the substance recognizing a membrane protein co-existing with PD-1 on a cell membrane if a suitable distance can be maintained. More specifically, it may be a peptide, amide, etc.
  • a linker can be used what is marketed, for example, Phenylenedimaleimide (manufactured by Aldrich) is available.
  • a substance that specifically recognizes PD-1 which is the subject of the present invention, can be produced as follows, for example.
  • hybrid antibody In the case that an antibody is chosen as a substance which recognizes PD-1 specifically, and that an antibody is also chosen as a substance which recognizes specifically a membrane protein co-existing with PD-1 on a cell membrane (hereinafter, simply referred to as “the membrane protein”)), a substance is called hybrid antibody by this invention. The method of producing this hybrid antibody is explained.
  • F(ab′) 2 the antibodies produced are treated with pepsin, isolated and purified, (9) Each F(ab′) 2 thus prepared is reduced, isolated and purified, (10) A hybrid antibody can be produced by bridging each F(ab′) SH thus prepared with a linker.
  • both or one of the two of a substance which specifically recognizes PD-1 and a substance which specifically recognizes a membrane protein co-existing with PD-1 on a cell membrane are a low molecule organic compound
  • a low molecule compound which inhibits the binding of each antibody to PD-1 or the membrane protein which is a corresponding antigen, respectively, is found out by using antibody produced by the above-mentioned technique and suitable detection equipment, (12) A substance can be produced by bridging the compound-to-compound, the compound to antibody, or the compound to Fab with a linker.
  • an immunization step (1) it is desirable to administer PD-1 or the membrane protein to an animal into the peritoneal cavity or footpad.
  • the animal to be immunized will not be limited especially if it is the one from which the monoclonal antibody is obtained generally such as mouse, rat or the like. In the case of a mouse, the amount of an antigen is enough if 10-200 micrograms is administered per time.
  • the cell fusion of (2) is carried out by excising spleen from an immunized animal with which the antibody titer has fully risen among the animals immunized in the step (1), preparing spleen cell suspension according to a usual method, and adding polyethylene glycol (preferably PEG4000) to a mixture of the spleen cells thus obtained and myeloma cells at 37° C.
  • polyethylene glycol preferably PEG4000
  • HGPRT hyperxanthine-guanine phosphoribosyl transferase-defective cell line which cannot survive in HAT medium (a medium containing hypoxanthine, aminopterin and thymidine)
  • HAT medium a medium containing hypoxanthine, aminopterin and thymidine
  • SP-2/O—Ag-14 is used.
  • the obtained cell fusion mixture is dispensed into a 96-well micro plate at a low density, and cultivated in the HAT medium.
  • HAT medium By culturing them for 1 to 2 weeks, un-fused myeloma cells, hybridomas of myeloma cells themselves, un-fused spleen cells and dybridomas of spleen cells themselves die because their surviving conditions are not satisfied, and only the hybridomas of spleen cell with myeloma cell are propagated.
  • a hybridoma is the one which produces an antibody against PD-1 or the membrane protein is judged by allowing each hybridoma culture supernatant to react with the immobilized antigen, and then determining amount of the antibody in the supernatant specifically adsorbed to the antigen using a labeled second antibody.
  • the step (4) is carried out by cloning the antibody-producing hybridoma in accordance with the soft agar culture method (Monoclonal Antibodies, 372 (1980)). In this case, it is also possible to use the limiting dilution method.
  • the step (5) is carried out by culturing the cloned hybridoma in usual medium and then separating and purifying from the culture supernatant, however, to obtain a larger amount of the antibody efficiently a method in which the hybridoma is administered into the abdominal cavity of mouse, allowed to propagate therein and then separated and purified from the ascites is used.
  • the purification can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, affinity chromatography using protein A-sepharose CL-4B (manufactured by Amersham Bioscience) is used more effectively.
  • a hybrid antibody of the present invention recognizes PD-1 specifically, it can be used for the purification and concentration of PD-1, for example, for affinity chromatography etc.
  • the step (7) can be carried out by bridging a linker such as sulfo-EMCS (N-(6-maleimidcaproxy) sulfo-succinimide sodium salt) to amide groups or SH (mercapto) groups of an antibody.
  • a linker such as sulfo-EMCS (N-(6-maleimidcaproxy) sulfo-succinimide sodium salt)
  • amide groups or SH (mercapto) groups of an antibody amide groups
  • one antibody is combined to sulfo-EMCS by amide coupling, un-reacted sulfo-EMCS is discarded by gel filtration, the maleimide groups of the sulfo-EMCS that is bound to the first antibody is reacted with SH (mercapto) groups of the other antibody that is reduced with 2-mercaptoethylamine etc., and then a substance bridged over two kinds of antibodies is size-fractionated using gel filtration.
  • SH mercap
  • the step (8) is carried out by digesting each antibody obtained in the step (6) with pepsin at 37° C. for 48 hours.
  • the separation and purification of F(ab′) 2 digested with pepsin can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 (manufactured by Amersham Bioscience) is used more effectively.
  • the step (9) is carried out by reducing F(ab′) 2 with 2-mercaptoethanol at 30° C. for 30 minutes.
  • the separation and purification of the reduced Fab SH can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 is used more effectively.
  • Fab SH fraction of one antibody is combined with a linker.
  • a linker a substance that is combinable with mercapto (SH) groups of Fab SH may be used, for example, a reaction is performed by adding phenylene dimaleimide for 30 minutes at room temperature. Next, the reaction is followed by adding the other Fab SH multiplied by 1.3 at room temperature for 4 hours.
  • the separation and purification of bispecific substance can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 is used more effectively.
  • the step (11) can be carried out by using the antibody obtained at the step (6) without modification, or by using the antibody with appropriate labeling (for example, biotin-conjugated or FITC-conjugated etc.) in accordance with a usual method.
  • an antigen is immobilized by a usual method, and then an antibody is added.
  • enzyme-conjugated second antibody and biotin-conjugated antibody are used, enzyme-conjugated streptavidin is added, then the specific binding between antigen and antibody is measured in the presence of chromophore-producing substance by using absorptiometer.
  • the obtained low molecule can be combined with the antibody or Fab by introducing the suitable functional group in it.
  • the suitable functional group for example, when maleimide groups is introduced, it is possible to make it combine with mercapto groups of an antibody or Fab.
  • both substances are low molecules, it is possible to synthesize a molecule containing both ones.
  • bispecific antibody in the present invention.
  • the method of producing this bispecific antibody is explained.
  • An antibody gene is isolated from hybridoma cells producing antibodies against PD-1 and the membrane protein, respectively, (2) A variable domain of an antibody gene against PD-1 and a variable domain of an antibody gene against the membrane protein are connected using a linker DNA, the connected DNA fragment is inserted to an expression vector, and cells are transfected with the expression vector and propagated, (3) Bispecific antibody can be prepared by separating and purifying the protein thus produced.
  • the step (1) consists of the processes, which isolates RNA from hybridoma cells, and which isolates cDNA encoding an antibody or its partial peptide.
  • RNA or mRNA from hybridoma cells can be carried out in accordance with known methods (hereinafter, if unstated especially, a known method is the method described in Molecular Cloning (Sambrook J., Fritsch E. F. and Maniatis T., Cold Spring Harbor Laboratory Press, 1989) or Current Protocol in Molecular Biology (Ausubel F. M. et al., John Wiley & Sons and Inc.).
  • the cloning of a cDNA encoding an antibody gene or its partial peptide of the present invention can be performed by an amplification using Polymerase Chain Reaction (hereinafter simply referred to as “PCR method”) with synthesized DNA primers having partial nucleotide sequence encoding an antibody of the present invention, or by a selection using hybridization of cDNAs inserted into a suitable vector with a labeled DNA fragment or synthetic DNA encoding an antibody of the present invention partially or entirely. Hybridization can be carried out in accordance with known methods.
  • An antibody gene may be amplified directly by using Reverse Transcriptase Polymerase Chain Reaction (hereinafter simply referred to as “RT-PCR method”) from total RNA or mRNA.
  • Bispecific antibody of the present invention may be prepared by:
  • Examples of expression system (host-vector system) for the production of peptide by using recombinant DNA technology are the expression systems using bacteria, yeast, insect cells and mammalian cells.
  • the initiation codon is added to 5′-end of the nucleotide sequence, for example, shown in SEQ ID NO:28, then the expression vector is prepared by connecting the obtained DNA to the downstream of a suitable promoter (e.g., trp promoter, lac promoter, ⁇ PL promoter, and T7 promoter), and by inserting it into a vector (e.g., pBR322, pUC18 and pUC19) which functions in an E. coli strain.
  • a suitable promoter e.g., trp promoter, lac promoter, ⁇ PL promoter, and T7 promoter
  • a vector e.g., pBR322, pUC18 and pUC19
  • an E. coli strain (e.g., E. coli strain DH1, E. coli strain JM109 and E. coli strain HB101) which is transformed with the expression vector described above may be cultured in an appropriate medium to obtain the desired peptide.
  • a signal peptide of bacteria e.g., signal peptide of pel B
  • the desired peptide may be released in periplasm.
  • a fusion protein with other peptide may be produced easily.
  • an expression vector is prepared by inserting a DNA, for example, having the nucleotide sequence shown in SEQ ID NO: 28, into the downstream of a proper promoter (e.g., SV40 promoter, LTR promoter and metallothionein promoter) in a proper vector (e.g., retro virus vector, papilloma virus vector, vaccinia virus vector and SV40 vector).
  • a proper promoter e.g., SV40 promoter, LTR promoter and metallothionein promoter
  • a proper vector e.g., retro virus vector, papilloma virus vector, vaccinia virus vector and SV40 vector.
  • suitable mammalian cells e.g., monkey COS-1 cells, COS-7 cells, Chinese hamster CHO cells, mouse L cells, 293 cells etc.
  • suitable mammalian cells e.g., monkey COS-1 cells, COS-7 cells, Chinese hamster CHO cells, mouse L cells, 293 cells etc.
  • the transfected cells are cultured in an appropriate medium, the aimed peptide can be secreted into the culture medium.
  • a transformation of E. coli can be carried out in accordance with the method, for example, described in Proc. Natl. Acad. Sci. (USA) 69:2110, 1972 and Gene 17: 107, 1982.
  • a transfection of mammalian cells can be carried out in accordance with the method, for example, described in Saiboukougaku supl. 8:263 (New experimental protocol for cell technology), Shujun-sha, 1995 and Virology 52:456, 1973.
  • a bispecific antibody can be prepared directly by using recombinant DNA technology.
  • Alt et al. FEBS Letter 454:90, 1999
  • VH heavy chain variable domain
  • VL light chain variable domain
  • VH and VL domains of this fragment are obliged to pair with each complementary VL and VH domain on other chain, and thereby form two antigen binding sites.
  • a peptide linker contains 3 to 12 amino acid residues, but it is not limited particularly to its amino acid sequence (Hudson et al., J. Immunol. Met. 231:177, 1999).
  • the peptide thus obtained can be purified by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography.
  • bispecific antibody of the present invention also recognizes PD-1 specifically, it can be used for the purification and concentration of PD-1, for example, for affinity chromatography etc.
  • the greatest and important usage of a substance that specifically recognizes PD-1 by the present invention is for the medical treatment of the following disease.
  • a substance that specifically recognizes PD-1 by the present invention is useful for the medical treatment and/or prevention of diseases such as neurodegenerative disease (Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease etc.).
  • diseases such as neurodegenerative disease (Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease etc.).
  • a substance that specifically recognizes PD-1 by the present invention is also useful for the medical treatment and/or prevention of diseases, in which PD-1 is involved and immune responses are enhanced, such as autoimmune diseases (glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis etc.), organ transplant rejection, and allergy.
  • autoimmune diseases glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lup
  • a substance that specifically recognizes PD-1 by the present invention is also useful for the medical treatment and/or prevention of diseases, in which PD-1 is involved and immune responses are reduced, such as neoplasm and infections.
  • administration of the substance that specifically recognizes PD-1 by the present invention can be carried out in systemic or local, generally peroral or parenteral ways.
  • the dosage to be administered depends upon age, body weight, symptom, desired therapeutic effect, route of administration, and duration of the treatment etc.
  • one dose per person is generally between 0.1 mg and 100 mg by oral administration up to several times per day, or between 0.01 mg and 30 mg by parenteral administration (preferably intravenous administration) up to several times per day, or continuous administration between 1 and 24 hrs. per day into vein.
  • the doses to be used depend upon various conditions. Therefore, there are cases in which doses lower than or greater than the ranges specified above may be used.
  • the compounds of the present invention may be administered as inner solid compositions or inner liquid compositions for oral administration, or as injections, liniments or suppositories etc. for parenteral administration.
  • Inner solid compositions for oral administration include compressed tablets, pills, capsules, dispersible powders and granules etc.
  • capsules include hard capsules and soft capsules.
  • one or more of the active compound(s) remains intact, or is/are admixed with excipients (lactose, mannitol, glucose, microcrystalline cellulose and starch etc.), connecting agents (hydroxypropyl cellulose, polyvinylpyrrolidone, magnesium metasilicate aluminate, etc.), disintegrating agents (cellulose calcium glycolate etc.), lubricating agents (magnesium stearate etc.), stabilizing agents, assisting agents for dissolving (glutamic acid, asparaginic acid etc.) etc. to prepare pharmaceuticals by known methods.
  • excipients lactose, mannitol, glucose, microcrystalline cellulose and starch etc.
  • connecting agents hydroxypropyl cellulose, polyvinylpyrrolidone, magnesium metasilicate aluminate, etc.
  • disintegrating agents cellulose calcium glycolate etc.
  • lubricating agents magnesium stearate etc.
  • stabilizing agents assisting agents for dissolving (glutamic acid
  • the pharmaceuticals may, if desired, be coated with coating agent (sugar, gelatin, hydroxypropyl cellulose or hydroxypropylmethyl cellulose phthalate etc.), or be coated with two or more films. Further, coating may include capsules of absorbable materials such as gelatin.
  • Inner liquid compositions for oral administration may contain pharmaceutically acceptable water-agents, suspensions, emulsions, syrups and elixirs etc.
  • one or more of the active compound(s) is/are resolved, suspended or emulsified in inert diluent (s) commonly used in the art (purified water, ethanol or mixture thereof etc.).
  • Such liquid compositions may also comprise wetting agents, suspending agents, emulsifying agent, sweetening agents, flavoring agents, perfuming agents, preserving agents and buffer agents etc.
  • Injections for parenteral administration include solutions, suspensions and emulsions and solid injections, which are dissolved or suspended in solvent when it is used.
  • one or more active compound(s) is/are dissolved, suspended or emulsified in a solvent.
  • Solvents include distilled water for injection, physiological salt solution, plant oil, propylene glycol, polyethylene glycol, alcohol such as ethanol, and mixture thereof etc.
  • Such compositions may comprise additional stabilizing agents, assisting agents for dissolving (glutamic acid, asparaginic acid, POLYSOLBATE80 (registered trademark) etc.), suspending agents, emulsifying agents, soothing agent, buffer agents, preserving agents etc.
  • They may be manufactured or prepared by sterilization or by aseptic manipulation in a final process. They may also be manufactured in the form of sterile solid compositions such as freeze-dried compositions, and can be dissolved in sterile water or some other sterile solvent for injection immediately before use.
  • compositions for parenteral administration include liquids for external use, ointments, endermic liniments, aerosols, spray compositions, suppositories and pessaries for vaginal administration etc., which comprise one or more of the active compound(s) and may be prepared by known methods.
  • Spray compositions may comprise additional substances other than inert diluents generally used: e.g. stabilizing agents such as sodium hydrogen sulfate, buffer agents to give isotonicity, isotonic buffer such as sodium chloride, sodium citrate and citric acid.
  • stabilizing agents such as sodium hydrogen sulfate, buffer agents to give isotonicity
  • isotonic buffer such as sodium chloride, sodium citrate and citric acid.
  • the substance that specifically recognizes PD-1 by the present invention can also be used for the screening of substances, which are involved in immune responses, by measuring the expression of PD-1.
  • a substance that specifically recognizes PD-1 by the present invention comprises a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker, is a superior substance recognizable both PD-1 and the membrane protein specifically and transmittable the signal of PD-1.
  • FIG. 1 shows the results of FACS analysis using anti-PD-1/anti-TCR hybrid Fab antibodies
  • FIG. 2 shows the effect of anti-PD-1/anti-TCR hybrid Fab antibodies on activated T cells
  • FIG. 3 shows the inhibitory effect of anti-PD-1/anti-BCR hybrid Fab antibodies on the production of IL-2 by anti-BCR antibody stimulation
  • FIG. 4 shows the effect of anti-PD-1/anti-BCR hybrid Fab antibodies on SHP-2 recruitment after anti-BCR antibody stimulation
  • FIG. 5 shows the plasmid J43-2C11scDb-pSec/hygro B for the expression of J43-2C11 bispecific antibody
  • FIG. 6 shows the effect of J43-2C11 bispecific antibodies on the production of IFN-r by activated mouse spleen T cells.
  • anti-PD-1 antibody and anti-T cell receptor antibody are linked by a linker simply referred to as “anti-PD-1/anti-TCR hybrid antibody”, and a substance in which anti-PD-1 antibody and anti-B cell receptor antibody are linked by a linker “anti-PD-1/anti-BCR hybrid antibody”, respectively.
  • Anti-mouse CD3 ⁇ monoclonal antibodies were substituted with Sodium phosphate (0.1M, pH7.0) and Nacl (50 mM), then 200 times quantity of sulfo-EMCS (manufactured by Doj in Chemical) were added and incubated at 20° C. for 1 hour. Then the reaction mixture was size-fractionated by gel filtration using Sephacril S-300 [Sodium phosphate (0.1M, pH7.0)] and major peak fractions were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • Antibodies against mouse PD-1 (produced by the hybridoma cells named (J43) and deposited as international deposition Accession number FERM BP-8118) were substituted with Sodium phosphate (0.1M, pH6.0), added with 2-mercaptoethylamine (final concentration of 10 mM) and EDTA (final concentration of 1 mM), and reduced at 37° C. for 90 minutes. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-300 [Sodium phosphate (0.1M, pH6.0)] and major peak fractions (single chain fractions) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • Antibodies against mouse PD-1 (produced by the hybridoma cells named (J43) and deposited as international deposition Accession number FERM BP-8118) were substituted with pepsin-buffer [(sodium acetate 0.1M, pH4.5), NaCl (0.1M)], added with pepsin (final concentration of 0.2 mg/ml), and digested for 48 hours at 37° C. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′) 2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • Anti-mouse CD3 ⁇ monoclonal antibodies (manufactured by Pharmingen) were substituted with pepsin-buffer [(sodium acetate 0.1M, pH4.5), NaCl (0.1M)], added with pepsin (final concentration of 0.2 mg/ml), and digested for 48 hours at 37° C. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′) 2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • Phenylenedimaleimide (manufactured by Aldrich) (final concentration of 4 mM) was added to the Fab SH fraction of anti-PD-1 antibodies prepared in step (2-A), and incubated for 30 minutes at room temperature to prepare J43 Fab mal fraction.
  • the J43 Fab mal fraction and the Fab SH fraction of anti-mouse CD3 ⁇ monoclonal antibodies were mixed at the rate of 1:1.3 and incubated for 4 hours at room temperature.
  • an appropriate amount of Tris-HCl (1M, pH8.0) were added to make the pH of the reaction mixture 8.0, 2-mercaptoethanol (final concentration of 20 mM) was added and incubated at 30° C. for 30 minutes.
  • iodoacetoamide (manufactured by Sigma) was added (final concentration of 25 mM) and incubated for additional 10 minutes at room temperature under light shielding.
  • reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (BsAb fraction) were collected by monitoring the absorbency at 280 nm.
  • the protein content was calculated from the absorbency at 280 nm simultaneously.
  • 1 ⁇ 10 6 cells were recovered from mPD-1/A20IIA1.6 cells as a PD-1 positive and CD3 negative cell, and naive T cells prepared from mouse spleen cells as a PD-1 negative and CD3 positive cell, respectively.
  • the cells were added with 91 ⁇ l of FACS buffer (PBS( ⁇ ) containing 0.5% BSA, EDTA (2 mM) and 0.01% NaN 3 ), 5 ⁇ l of mouse serum and 4 ⁇ l of hybrid antibodies (2 ⁇ g) and incubated for 30 minutes on ice. After washing with PBS( ⁇ ) once, the cells were added with each 2 ⁇ l (1 ⁇ g) of second antibodies, fill upped to final 100 ⁇ l with FACS buffer, and incubated for 30 minutes on ice. Then they were analyzed by using FACScan. The results were shown in FIG. 1 (in the following figures, hybrid Fab antibody simply referred to as “HFAb”).
  • Spleen was excised from BALB/c mouse. After red blood cells were hemolyzed, the spleen cells were washed once with PBS ( ⁇ ) and suspended in medium RPMI 1640 (10% FCS, antibiotics) (1 ⁇ 10 8 cells/ml). Next, T cells were prepared by using nylon fiber column (manufactured by WAKO) for T cell separation equilibrated with the medium.
  • Antibodies against mouse PD-1 were substituted with pepsin-buffer [(sodium acetate 0.1M, pH4.5), NaCl (0.1M)], added with pepsin (manufactured by SIGMA) (final concentration of 0.2 mg/ml), and digested at 37° C. for 48 hours. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 (manufactured by AmarshamParmacia) [Tris-HCl (0.2M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′) 2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • Phenylenedimaleimide (manufactured by Aldrich) (final concentration of 4 mM) was added to the Fab SH fraction of J43 anti-PD-1 antibodies, and incubated for 30 minutes at room temperature to prepare J43 Fab mal fraction.
  • the J43 Fab mal fraction and the Fab SH fraction of anti-IgG (H+L) polyclonal antibodies were mixed at the rate of 1:1.3 and incubated for 4 hours at room temperature.
  • an appropriate amount of Tris-HCl (1M, pH8.0) were added to make the pH of the reaction mixture 8.0, 2-mercaptoethanol (final concentration of 20 mM) was added and incubated at 30° C. for 30 minutes.
  • DNA fragments of mPD1-flag digested with EcoRI were inserted into the EcoRI site of a commercially available expression vector to construct the expression plasmid mPD1-pA.
  • the cells were suspended in 30 ml of medium (RPMI1640 containing 10% FBS, 50 ⁇ l of 2-mercaptoethanol, penicillin and streptomycin), diluted thirty fold further, and dispensed onto 96-well plates (103/100 ⁇ l/well). After 48 hours, the selection was initiated using final 3 ⁇ M of Puromycin to establish the cell line expressing mouse PD-1.
  • medium RPMI1640 containing 10% FBS, 50 ⁇ l of 2-mercaptoethanol, penicillin and streptomycin
  • A20IIA1.6 cells that was forced to express mouse PD-1 were seeded on 96-well plates (5 ⁇ 10 5 cells/100 ⁇ l).
  • Anti-PD-1/anti-BCR hybrid Fab antibodies (0, 1, 3, 10 ⁇ g/100 ⁇ l) were added, 10 minutes later 100 ⁇ l of anti-mouse IgG (H+L) F(ab′) 2 (manufactured by Zymed) (final concentration of 0.3, 1, 3 ⁇ g/ml) were dispensed and cultivated for 12 hours in a CO 2 incubator (at 37° C.).
  • the culture supernatants were recovered and the concentrations of IL-2 in the culture supernatants were measured by using mouse IL-2 assay kit (manufactured by R & G System). The results were shown in FIG. 3 .
  • Anti-PD-1/anti-BCR hybrid Fab antibodies (0, 1, 3, 10 ⁇ g/100 ⁇ l) were added to 3 ⁇ 10 6 of A20IIA1.6 cells (B cell line) that was forced to express mouse PD-1. After 10 minutes, 100 ⁇ l of anti-mouse IgG (H+L) F(ab′) 2 were added and incubated for 5 minutes at room temperature.
  • the cells were suspended in 200 ⁇ l of lysis buffer (composition: Tris-HCl (20 mM, pH7.4), NaCl (150 mM), Na 2 EDTA (1 mM), EGTA (1 mM), 1% Triton-X100, sodium pyrophosphate (2.5 mM), 1-sodium glycerophosphate (1 mM), Na 3 VO 4 (1 mM), leupeptin (1 ⁇ g/ml) and PMSF (1 mM) and left at rest on ice.
  • lysis buffer composition: Tris-HCl (20 mM, pH7.4), NaCl (150 mM), Na 2 EDTA (1 mM), EGTA (1 mM), 1% Triton-X100, sodium pyrophosphate (2.5 mM), 1-sodium glycerophosphate (1 mM), Na 3 VO 4 (1 mM), leupeptin (1 ⁇ g/ml) and PMSF (1 mM) and left at rest
  • the beads were washed five times with 400 ⁇ l of lysis buffer, added with 20 ⁇ l of lysis buffer and 20 ⁇ l of 2 ⁇ SDS sample buffer, and boiled at 100° C. for 5 minutes. After discarding the beads by centrifugation, 15 ⁇ l of supernatants were subjected to 4-20% SDS-PAGE. After electrophoresis the gels were substituted with blotting buffer and transferred onto PVDF membrane (manufactured by BIO RAD). Then the membrane was blocked with Block Ace (manufactured by Dainippon Pharmaceuticals) for 1 hour at room temperature.
  • Block Ace manufactured by Dainippon Pharmaceuticals
  • the membrane was incubated with anti-SHP-2 antibody (manufactured by SANTA CRUZ) diluted 1/200 for 1 hour at room temperature, then washed three times with TBS-T for 10 minutes.
  • the membrane was incubated with HRP-conjugated anti-rabbit Ig antibody (manufactured by Amersham Bioscience) diluted 1/2000 for 1 hour at room temperature, then washed three times with TBS-T for 10 minutes.
  • the membrane was emitted light by using ECT plus detection kit (manufactured by Amersham Bioscience) and analyzed using luminoimager LAS1000 plus (manufactured by FUJI Film).
  • Anti-mouse PD-1 antibody producing hybridoma (J43) cells were cultured in Hybridoma SFM medium (manufactured by Invitrogen) at 37° C. under 5% CO 2 , a few days after the culture supernatants of hybridoma cells were recovered. IgG fraction was purified from the culture supernatants recovered by using HiTrap Protein G (manufactured by Amersham Bioscience).
  • J43 IgG was subjected to 10-20% SDS-PAGE. After electrophoresis the IgG was electrically transferred from the gel onto PVDF membrane (manufactured by BIO RAD). The membrane transferred was stained with coomassie, the membrane fraction containing the light chain of J43 IgG was removed, and amino terminal 15 residues of the light chain were determined by using peptide sequencer Procise492 (manufactured by Applied Biosystems) (SEQ ID NO:1).
  • Degenerated primer (primer No. 1) was designed based on the amino terminal sequence (YELTQPPSASVNVGE) of the light chain determined by peptide sequencing.
  • 3′RACE was carried out by using 3′-Full RACE Core Set (manufactured by Takara Shuzo) under the following conditions.
  • Primer No.1 (SEQ ID NO: 2) 5′-ta(c/t) ga(a/g) ct(g/a/t/c) ac(g/a/t/c) ca(a/g) cc(g/a/t/c) cc-3′
  • 5′RACE 5′-Full RACE Core Set (manufactured by Takara Shuzo) under the following conditions.
  • Primer No.2 5′-ccc aag agg tca gga gtt gga-3′ (SEQ ID NO: 5) (5′ phosphorylated)
  • Primer No.3 5′-ttg acc agg cat ccc agg gtc-3′ (SEQ ID NO: 6)
  • Primer No.4 5′-cgt aag ctg gaa ctc tgg agc-3′ (SEQ ID NO: 7)
  • Primer No.5 5′-tgg ttg tgc tgt cac agg cag-3′ (SEQ ID NO: 8)
  • Primer No.6 5′-tgc aca cct tcc cat ctg tcc t-3′ (SEQ ID NO: 9)
  • RNA (ssDNA) Ligation buffer 8 40% Polyethylene glycol (PEG) #6000 20 d H 2 O 12 Pellet after ethanol precipitation T4 RNA ligase 1 41 ⁇ l 15° C. 15 hours
  • Anti-mouse CD3 ⁇ antibody producing hybridoma 145-2C11: manufactured by Pharmingen cells were cultured in Hybridoma SFM medium (manufactured by Invitrogen) at 37° C. under 5% CO 2 , a few days after 5 ⁇ 10 6 hybrisoma cells were lysed with 1 ml of TRIzol (manufactured by Invitrogen). Total RNA was prepared according to the direction of attached document.
  • cDNA was synthesized from 2.5 ⁇ g of total RNA extracted from hybridoma (145-2C11) cells by oligo-dT prime method using Ready-To-Go You-Prime First-Strand Beads (manufactured by Amersham Pharmacia). Operations and procedures were followed by the instructions of attached document.
  • primers No. 7 and No. 8 were designed.
  • primers No. 9 and No. 10 were designed. PCR was carried out using these primers and the cDNA library from hybridoma 145-2C11 as a template.
  • Primer No.7 (SEQ ID NO: 12) 5′-gag gtg cag ctg gtg gag tct-3′
  • Primer No.8 (SEQ ID NO: 13) 5′-tga gga gac ggt gac cat ggt t-3′
  • Primer No.9 (SEQ ID NO: 14) 5′-gac atc cag atg acc cag tct c-3′
  • Primer No.10 (SEQ ID NO: 15) 5′-ttt gat ttc cag ctt ggt gcc ag-3′
  • the PCR products were subjected to electrophoresis using 1% Agarose gel, then the gel was stained by EtBr.
  • DNA fragments were recovered from the gel by using MinElute Gel Extraction Kit (manufactured by Quiagen), the DNA fragments recovered was ligated to pGEM-T Easy Vector (manufactured by Promega) by using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo).
  • E. coli DH5 ⁇ were transformed with the ligated plasmids.
  • the plasmids were purified from E. coli , and both DNA were sequenced. It was confirmed that their sequences were identical to those of GenBank Accession No. AF00357 and GenBank Accession No. AF000356, respectively.
  • J43 IgG heavy chain cDNA and 145-2C11 IgG light chain cDNA were connected by PCR using linker No. 1, No. 2, primer No. 11 and No. 12 to prepare fragment 1 (see FIG. 5 ).
  • 145-2C11 IgG light chain cDNA and 145-2C11 IgG heavy chain cDNA were connected by PCR using linker No. 3, No. 4, primer No. 13 and No. 14 to prepare fragment 2 (see FIG. 5 ).
  • 145-2C11 IgG heavy chain cDNA and J43 IgG light chain cDNA were connected by PCR using linker No. 5, No. 6, primer No. 15 and No. 16 to prepare fragment 3 (see FIG. 5 ).
  • Primer No.11 (SEQ ID NO: 16) 5′-ttt gaa ttc aga ggt gcg gct tct gga gtc t-3′ Primer No.12 (SEQ ID NO: 17) 5′-gat cag gag ctt agg agc ttt cc-3′ Primer No.13 (SEQ ID NO: 18) 5′-cag gcc agt cag gac att agc aa-3′ Primer No.14 (SEQ ID NO: 19) 5′-taa tgt atg cga ccg act cca gc-3′ Primer No.15 (SEQ ID NO: 20) 5′-tga ggc ctc tgg att cac ctt ca-3′ Primer No.16 (SEQ ID NO: 21) 5′-aaa aaaaaa ctc gag gac cta gg
  • the DNA fragments 1, 2 and 3 and plasmid pBluescriptII SK(+) (manufactured by StrateGene) were digested with restriction enzymes EcoRI/KpnI, KpnI/SphI, SphI/XhoI, EcoRI/XhoI, respectively. After electrophoresis on 1% agarose gel, the DNA fragments were purified from the gel using MinElute Gel Extraction Kit. Next, these three fragments and plasmid pBluescriptII SK(+) (manufactured by StrateGene) were connected using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo), then E. coli DH5 (were transformed with the connected plasmid.
  • Plasmid J43-2C11scDb-pBluescriptII SK(+) was prepared from E. coli , then the nucleotide sequence of the insert was determined (SEQ ID NO:28). The deduced amino acid sequence is shown in sequence listing (SEQ ID NO:29).
  • J43-2CllscDb-pBluescriptII SK(+) was digested with restriction enzymes BamHI and XhoI, the BamHI-XhoI fragment out of BamHI-XhoI and BamHI-BamHI fragments generated by the digestion was connected with BamHI/XhoI digested mammalian expression vector pSecTag2/Hygro B (manufactured by Invitrogen). Next, the pSecTag2/Hygro B connected with BamHI-XhoI fragment was digested again with restriction enzyme BamHI, and connected with the other BamHI-BamHI fragment. E. coli DH5 ⁇ were transformed with the connected plasmid.
  • Plasmid J43-2C11scDb-pSec/Hygro B was prepared from E. coli , then the nucleotide sequence of thus prepared J43-2C11 bispecific antibody was determined (SEQ ID NO:30). The deduced amino acid sequence is shown in sequence listing (SEQ ID NO:31).
  • T cells 6 ⁇ 10 6 293 T cells were suspended in 20 ml of medium (DMEM containing 10% FBS) and seeded into 150 mm dish coated with Type Ic collagen. Next day, the cells were washed with 10 ml of DMEM and transfected with J43-2C11scDb-pSec/Hygro B by using LipofectAMINE-plus. After 3 hours, 5 ml of DMEM containing 40% FBS were added to the cells. At day 2, the cells were washed with 10 ml of DMEM, added 20 ml of new DMEM, and the culture supernatant was recovered at day 4.
  • medium DMEM containing 10% FBS
  • the cells were discarded by centrifugation, then the supernatant was filtrated using 0.22 ⁇ m PVDF filter.
  • the supernatant was enclosed in dialysis tube, dialyzed against PBS containing 40% PEG20000, and concentrated.
  • the concentrated supernatant was purified using HiTrap chelating HP column (manufactured by Amersham Farmacia).
  • the antibodies were purified by gel-filtration using Hiprep 16/60 Sephacryl S-200 High Resolution (manufactured by Amersham Farmacia).
  • Spleen was removed from BALB/c mouse, and cells were prepared using CellStrainer (70 ⁇ m Nyron). The cells were recovered by centrifugation, and red blood cells were hemolyzed by the addition of hemolysis buffer [NH 4 Cl (0.8%), KCO 3 (0.1%) and EDTA (1 mM)]. The cells were washed with PBS ( ⁇ ) once, T cells were enriched by using mouse CD3 + T cell enrichment column kit (manufactured by R&D), and suspended in medium (RPMI1640 containing 10% FBS) in the proportion of 5 ⁇ 10 6 cells/ml.
  • the T cells thus prepared were seeded on 96-well plates (2 ⁇ 10 5 cells/100 ⁇ l/well), which were coated preliminarily with 5 ⁇ g/ml of anti-CD3 antibodies (clone KT3) at 37° C. for 3 hours, added with J43-2C11 bispecific antibody diluted in the medium (0.01, 0.03, 0.1, 0.3, 1 and 3 ⁇ g/100 ⁇ l), and cultivated for 72 hours at 37° C. under 5% CO 2 . After 72 hours, the culture supernatants were recovered, and the concentrations of IFN-r in the supernatants were determined by using Quantikine Immunoassay Kit (manufactured by R&D).
  • J43-2C11 bispecific antibody suppressed dose dependently the production of IFN-r from activated mouse spleen T cells in vitro.

Abstract

Compounds that can recognize selectively both PD-1 protein and a membrane protein co-existing with PD-1 on a cell membrane, and can transmit a suppressive signal of PD-1. The compounds are useful for medical treatment and/or prevention of diseases caused by immune abnormality.

Description

  • This is a divisional of application Ser. No. 10/485,466 filed Feb. 2, 2004, which is the National Stage of PCT/JP02/07735 filed Jul. 30, 2002, and which claims priority from Japanese Application No. 2001-232303 filed Jul. 31, 2001. The entire disclosures of the prior applications are hereby incorporated by reference.
  • TECHNICAL FIELD
  • The invention is related to a substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker.
  • BACKGROUND
  • The immune system acquired the mechanism that can respond to various foreign antigens. The mechanism brings about the diversity of an antigen receptor by recombination of V, (D) and J fragment in T cells and B cells. Although this mechanism brought a result that produces self-reactive lymphocytes simultaneously, these self-reactive lymphocytes are removed by the negative selection in the thymus or bone marrow, and are further controlled by the self-tolerance mechanism of clone removal or anergy in the periphery.
  • Although it is thought that an autoimmune disease is developed by the breakdown of self-tolerance, the researches using various disease model mice have been made towards the elucidation of the mechanism of pathogenesis of the disease. However, there are many still unknown matters about the etiology of an autoimmune disease and the molecular mechanism of self-tolerance. In such a situation, existence of the mouse which shows the symptoms of an autoimmune disease with a single gene deficit is very important, when trying to study the etiology of an autoimmune disease from a molecular biological viewpoint. CTLA4−/−mouse which causes lethal systemic lymphocytes infiltration (Waterhouse P. et. al., Science 270:985˜988, 1995, Tivol E. A. et. al., Immunity 3:541˜547, 1995), SHP-1 deficit mothaten mice (Shulltz L. D. et. al., Cell 73: 1445˜1454, 1993), TGF-beta-1 knockout mouse (Shull M. M. et. al., Nature 359:693-699, 1992), lyn−/−mouse which shows the symptoms of glomerular nephritis (Hibbs M. L. et. al., Cell 83:301-311, 1995), and FCRIIB−/−mouse (Bolland S. & Ravetch J. V., Immunity 13:277-285, 2000) are the representation, and the relation of these molecules and self-tolerance is studied.
  • PD-1 is type I membrane protein of 55 kD belonging to an immunoglobulin family. Bothmouse PD-1 and human PD-1 consist of 288 amino acids, and have signal peptide at N terminal (20 amino acid) and hydrophobic region in the middle part, which is a transmembrane region (The EMBO J. 11 (11):3887-3895, 1992); Japanese patent Publication No. 5-336973; EMBL/GenBank/DDJB Acc. No. X67914, Genomics 23:704, 1994; Japanese patent Publication No. 7-291996 (U.S. Pat. No. 5,629,204).
  • In a thymocyte, PD-1 is expressed during a CD4−CD8− cell differentiation to a CD4+ CD8+ cell (Nishimura H. et. al., Int. Immunol. 8:773-780, 1996, Nishimura H. et. al., J. Exp. Med. 191:891-898, 2000). Moreover, in the periphery, PD-1 is expressed in T cells and B cells which were activated by the stimulus from an antigen receptor (Agata Y. et. al., Int. Immunol. 8:765-772, 1996), and in bone marrow cells including activated macrophage.
  • PD-1 has ITIM (Immunoreceptor tyrosine-based inhibitory motif) in its intracellular region, therefore it is considered to be a negative regulator in immune reaction. PD-1 deficit mice develop a lupus-like autoimmune disease such as glomerular nephritis and arthritis (C57BL/6 genetic background) (Nishimura H. et. al., Int. Imuunol. 10:1563-1572, 1998, Nishimura H. et. al., Immunity 11:141-151, 1999) and dilated cardiomyopathy-like disease (BALB/c genetic background) (Nishimura H. et. al., Science 291:319-322, 2001), it became clear that PD-1 is a regulator of the development of autoimmune disease, especially one of self-tolerance in the periphery.
  • DISCLOSURE OF THE INVENTION
  • It is thought that PD-1 is the regulator of various autoimmune diseases, and that it is one of the genes that cause an autoimmune disease. By controlling the function of PD-1, it thought that the medical treatment and diagnosis of suppression or enhancement of the immune function, infection, the rejection at the time of a transplant, a neoplasm, etc. could be performed, and as a result of repeating examination wholeheartedly, the inventors reached this invention concerning the substance which controls the function of PD-1.
  • The stimulus to lymphocytes which control immunity is transmitted mainly through T cell receptor (TCR) in the case of T cells, and B cell receptor (BCR) in the case of B cells, and the intracellular phosphorylation play an important role in its molecular mechanism.
  • Since it became clear that PD-1 is controlling negatively various cells responsible for immunity, such as lymphocytes and myeloid cells etc., and PD-1 has ITIM (Immunoreceptor tyrosine-based inhibitory motif) in its intracellular region, the inventors considered the molecular mechanism involved in the inhibitory signal transduction of PD-1 to be the recruit of de-phosphorylation enzymes (phosphatases). Therefore, it came to be considered by locating PD-1 near TCR or BCR that it can display the function of PD-1. The inventors confirmed that the inhibitory signal of PD-1 was transmitted with the substance in which PD-1 was cross-linked to TCR or BCR, and completed this invention.
  • The inventors confirmed first that the above-mentioned idea was right using anti-PD-1 antibody, and anti-BCR antibody or anti-CD3 antibody. CD3 is the membrane protein expressed on a T cell, and is one component of the complexes that constitute TCR. The divalent antibody was constructed by bridging anti-PD-1 antibody and anti-BCR or anti-CD3 antibody. In the present invention, this divalent antibody is called hybrid antibody. The inventors produced this hybrid antibody for the first time.
  • Moreover, the knowledge that a signal transmits by cross-linking two kind of receptors using this hybrid antibody is also acquired for the first time.
  • Namely, the present invention relates to,
  • 1. A substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker,
    2. The substance according to (1), which is a divalent substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker,
    3. The substance according to (1) or (2), in which a membrane protein is a protein existing on a T cell membrane or B cell membrane,
    4. The substance according to (1) or (2), which comprises a substance that recognizes PD-1, a substance that recognizes a protein constituting T cell receptor complex or a substance that recognizes a protein constituting B cell receptor complex, and a linker,
    5. The substance according to any one of (1) to (4), in which a substance that recognizes PD-1 and in which a substance that recognizes a protein is dimer to pentamer, respectively,
    6. The substance according to any one of (1) to (5), in which one of or both substance that recognizes PD-1 and substance that recognizes a protein are an antibody,
    7. The substance according to any one of (1) to (6), in which one of the two or both substance that recognizes PD-1 and substance that recognizes a protein are a Fab portion of antibody,
    8. The substance according to any one of (1) to (5), in which a linker comprises an organic compound,
    9. The substance according to any one of (1) to (5), in which a linker comprises a peptide,
    10. The substance according to any one of (1) to (5), in which a substance that recognizes PD-1 and in which a substance that recognizes a protein is a peptide, respectively,
    11. The substance according to any one of (1) to (5), in which a substance that recognizes PD-1 and in which a substance that recognizes a protein comprises two or more peptides including a heavy chain variable region and a light chain variable region of antibody, respectively,
    12. The substance according to any one of (1) to (5), in which a substance that recognizes PD-1, in which a substance that recognizes a protein, and in which a linker is a peptide, respectively,
    13. A pharmaceutical composition containing an effective dose of the substance according to (1) for the medical treatment and/or prevention of a disease in which PD-1 participates,
    14. The pharmaceutical composition according to (13), in which disease is selected from the group consisting of neurodegenerative disease, autoimmune disease, organ transplant rejection, neoplasm and infection,
    15. The pharmaceutical composition according to (14), in which neurodegenerative disease is selected from the group consisting of Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease,
    16. The pharmaceutical composition according to (14), in which autoimmune disease is selected from the group consisting of glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis.
  • A substance that recognizes PD-1 said by the present invention may be a substance which recognizes PD-1 specifically, for example, anti-PD-1 antibody, the fragment of anti-PD-1 antibody, PD-1 in itself, the fragment of PD-1, ligand of PD-1 (PD-L1 (Freeman G. J. et. al., J. Exp. Med. 192:1027-1034, 2000), PD-L2, PD-H3), the fragment thereof and a low molecule organic compound, etc.
  • In more concretely, it is anti-PD-1 antibody produced by the hybridoma strain “J43”, which was deposited on May 30, 2001 in International Patent Organism Depositary, National Institute of Advanced Industrial Science and Technology, AIST Tsukuba Central 6, 1-1, Higashi 1-Chome Tsukuba-shi, Ibaraki-ken 305-8566 Japan as Accession number FERM P-18356, and transferred to international deposition on Jul. 16, 2002 as international deposition Accession number FERM BP-8118.
  • Preferably, although it is the Fab portion of the antibody, it is not limited to this.
  • A substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane said by the present invention may be a substance which recognizes a membrane protein specifically, for example, a substance which specifically recognizes a complex that constitute T cell receptor (TCR) (TCR complex) etc. expressed on T cells, or a substance which specifically recognizes a complex that constitute B cell receptor (BCR) (BCR complex) etc. expressed on B cells, such as the fragment of a protein constituting TCR complex, anti-TCR antibody, the fragment of anti-TCR antibody, the fragment of a protein constituting BCR complex, anti-BCR antibody and the fragment of anti-BCR antibody.
  • Preferably, although it is the Fab portion of an anti-TCR antibody or the Fab portion of an anti-BCR antibody, it is not limited to these.
  • Both an anti-TCR antibody and an anti-BCR antibody are available commercially. For example, anti-CD3 antibody (α-CD3ε mAb, manufactured by Pharmingen) as an anti-TCR antibody, and anti-IgG (H+L) polyclonal antibody (manufactured by Zymed) as an anti-BCR antibody are available, respectively.
  • A linker said by the present invention may be a substance which can connect the above mentioned substance recognizing PD-1 and the substance recognizing a membrane protein co-existing with PD-1 on a cell membrane if a suitable distance can be maintained. More specifically, it may be a peptide, amide, etc.
  • A linker can be used what is marketed, for example, Phenylenedimaleimide (manufactured by Aldrich) is available.
  • A substance that specifically recognizes PD-1, which is the subject of the present invention, can be produced as follows, for example.
  • In the case that an antibody is chosen as a substance which recognizes PD-1 specifically, and that an antibody is also chosen as a substance which recognizes specifically a membrane protein co-existing with PD-1 on a cell membrane (hereinafter, simply referred to as “the membrane protein”)), a substance is called hybrid antibody by this invention. The method of producing this hybrid antibody is explained.
  • (1) Animals are immunized with PD-1 or the membrane protein as an antigen,
    (2) Spleen cells from the immunized animal and myeloma cells are fused,
    (3) Cells producing monoclonal antibody against the antigen (PD-1 or the membrane protein) are screened among thus obtained hybridoma cells,
    (4) Aimed hybridoma cells are cloned,
    (5) Cloned hybridoma cells are grown,
    (6) Antibodies produced are isolated and purified,
    (7) A hybrid antibody can be produced by bridging thus obtained anti-PD-1 antibody and anti-membrane antibody with a linker.
  • Alternatively,
  • (8) To obtain F(ab′)2, the antibodies produced are treated with pepsin, isolated and purified,
    (9) Each F(ab′)2 thus prepared is reduced, isolated and purified,
    (10) A hybrid antibody can be produced by bridging each F(ab′)SH thus prepared with a linker.
  • In the case that both or one of the two of a substance which specifically recognizes PD-1 and a substance which specifically recognizes a membrane protein co-existing with PD-1 on a cell membrane (the membrane protein) are a low molecule organic compound,
  • (11) A low molecule compound which inhibits the binding of each antibody to PD-1 or the membrane protein which is a corresponding antigen, respectively, is found out by using antibody produced by the above-mentioned technique and suitable detection equipment,
    (12) A substance can be produced by bridging the compound-to-compound, the compound to antibody, or the compound to Fab with a linker.
  • It is as follows when each step is explained more concretely.
  • In an immunization step (1), it is desirable to administer PD-1 or the membrane protein to an animal into the peritoneal cavity or footpad. Moreover, the animal to be immunized will not be limited especially if it is the one from which the monoclonal antibody is obtained generally such as mouse, rat or the like. In the case of a mouse, the amount of an antigen is enough if 10-200 micrograms is administered per time.
  • The cell fusion of (2) is carried out by excising spleen from an immunized animal with which the antibody titer has fully risen among the animals immunized in the step (1), preparing spleen cell suspension according to a usual method, and adding polyethylene glycol (preferably PEG4000) to a mixture of the spleen cells thus obtained and myeloma cells at 37° C. Some kinds, such as P3X63Ag8, P3/NS1/1-Ag4-1, and SP-2/0-Ag-14, are known as the mouse myeloma cell, and all of them are easily available.
  • As the myeloma cell, HGPRT (hypoxanthine-guanine phosphoribosyl transferase)-defective cell line which cannot survive in HAT medium (a medium containing hypoxanthine, aminopterin and thymidine) is useful, and it is further preferred that it is a cell line in which the myeloma cells themselves do not secrete any antibody. Preferably, SP-2/O—Ag-14 is used.
  • Next, the obtained cell fusion mixture is dispensed into a 96-well micro plate at a low density, and cultivated in the HAT medium. By culturing them for 1 to 2 weeks, un-fused myeloma cells, hybridomas of myeloma cells themselves, un-fused spleen cells and dybridomas of spleen cells themselves die because their surviving conditions are not satisfied, and only the hybridomas of spleen cell with myeloma cell are propagated.
  • In the screening of (3), whether or not a hybridoma is the one which produces an antibody against PD-1 or the membrane protein is judged by allowing each hybridoma culture supernatant to react with the immobilized antigen, and then determining amount of the antibody in the supernatant specifically adsorbed to the antigen using a labeled second antibody.
  • The step (4) is carried out by cloning the antibody-producing hybridoma in accordance with the soft agar culture method (Monoclonal Antibodies, 372 (1980)). In this case, it is also possible to use the limiting dilution method.
  • The step (5) is carried out by culturing the cloned hybridoma in usual medium and then separating and purifying from the culture supernatant, however, to obtain a larger amount of the antibody efficiently a method in which the hybridoma is administered into the abdominal cavity of mouse, allowed to propagate therein and then separated and purified from the ascites is used.
  • In the step (6), the purification can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, affinity chromatography using protein A-sepharose CL-4B (manufactured by Amersham Bioscience) is used more effectively.
  • Since a hybrid antibody of the present invention recognizes PD-1 specifically, it can be used for the purification and concentration of PD-1, for example, for affinity chromatography etc.
  • The step (7) can be carried out by bridging a linker such as sulfo-EMCS (N-(6-maleimidcaproxy) sulfo-succinimide sodium salt) to amide groups or SH (mercapto) groups of an antibody. First, one antibody is combined to sulfo-EMCS by amide coupling, un-reacted sulfo-EMCS is discarded by gel filtration, the maleimide groups of the sulfo-EMCS that is bound to the first antibody is reacted with SH (mercapto) groups of the other antibody that is reduced with 2-mercaptoethylamine etc., and then a substance bridged over two kinds of antibodies is size-fractionated using gel filtration.
  • The step (8) is carried out by digesting each antibody obtained in the step (6) with pepsin at 37° C. for 48 hours. The separation and purification of F(ab′)2 digested with pepsin can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 (manufactured by Amersham Bioscience) is used more effectively.
  • The step (9) is carried out by reducing F(ab′)2 with 2-mercaptoethanol at 30° C. for 30 minutes. The separation and purification of the reduced FabSH can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 is used more effectively.
  • In the step (10), FabSH fraction of one antibody is combined with a linker. As a linker, a substance that is combinable with mercapto (SH) groups of FabSH may be used, for example, a reaction is performed by adding phenylene dimaleimide for 30 minutes at room temperature. Next, the reaction is followed by adding the other FabSH multiplied by 1.3 at room temperature for 4 hours. The separation and purification of bispecific substance can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 is used more effectively.
  • The step (11) can be carried out by using the antibody obtained at the step (6) without modification, or by using the antibody with appropriate labeling (for example, biotin-conjugated or FITC-conjugated etc.) in accordance with a usual method. In the case that ELISA method is used, an antigen is immobilized by a usual method, and then an antibody is added. Next, when enzyme-conjugated second antibody and biotin-conjugated antibody are used, enzyme-conjugated streptavidin is added, then the specific binding between antigen and antibody is measured in the presence of chromophore-producing substance by using absorptiometer. By using this assay system a low molecule that specifically recognizes PD-1 or the membrane protein can be obtained.
  • In the step (12), when one of the two is an antibody or Fab, the obtained low molecule can be combined with the antibody or Fab by introducing the suitable functional group in it. For example, when maleimide groups is introduced, it is possible to make it combine with mercapto groups of an antibody or Fab. Moreover, when both substances are low molecules, it is possible to synthesize a molecule containing both ones.
  • In the case that an antibody is chosen as a substance which recognizes PD-1 specifically, that an antibody is also chosen as a substance which recognizes specifically a membrane protein co-existing with PD-1 on a cell membrane, and that both antibodies are included in the same peptide, the substance is called bispecific antibody in the present invention. The method of producing this bispecific antibody is explained.
  • (1) An antibody gene is isolated from hybridoma cells producing antibodies against PD-1 and the membrane protein, respectively,
    (2) A variable domain of an antibody gene against PD-1 and a variable domain of an antibody gene against the membrane protein are connected using a linker DNA, the connected DNA fragment is inserted to an expression vector, and cells are transfected with the expression vector and propagated,
    (3) Bispecific antibody can be prepared by separating and purifying the protein thus produced.
  • It is as follows when each step is explained more concretely.
  • The step (1) consists of the processes, which isolates RNA from hybridoma cells, and which isolates cDNA encoding an antibody or its partial peptide.
  • The process which isolates total RNA or mRNA from hybridoma cells can be carried out in accordance with known methods (hereinafter, if unstated especially, a known method is the method described in Molecular Cloning (Sambrook J., Fritsch E. F. and Maniatis T., Cold Spring Harbor Laboratory Press, 1989) or Current Protocol in Molecular Biology (Ausubel F. M. et al., John Wiley & Sons and Inc.).
  • The cloning of a cDNA encoding an antibody gene or its partial peptide of the present invention can be performed by an amplification using Polymerase Chain Reaction (hereinafter simply referred to as “PCR method”) with synthesized DNA primers having partial nucleotide sequence encoding an antibody of the present invention, or by a selection using hybridization of cDNAs inserted into a suitable vector with a labeled DNA fragment or synthetic DNA encoding an antibody of the present invention partially or entirely. Hybridization can be carried out in accordance with known methods. An antibody gene may be amplified directly by using Reverse Transcriptase Polymerase Chain Reaction (hereinafter simply referred to as “RT-PCR method”) from total RNA or mRNA.
  • (2) Bispecific antibody of the present invention may be prepared by:
  • i) A method of peptide synthesis, or
    ii) A method of production using recombinant DNA technology, preferably, by the method described in ii) in an industrial production.
  • Examples of expression system (host-vector system) for the production of peptide by using recombinant DNA technology are the expression systems using bacteria, yeast, insect cells and mammalian cells.
  • For example, in an E. Coli expression system, the initiation codon (ATG) is added to 5′-end of the nucleotide sequence, for example, shown in SEQ ID NO:28, then the expression vector is prepared by connecting the obtained DNA to the downstream of a suitable promoter (e.g., trp promoter, lac promoter, λ PL promoter, and T7 promoter), and by inserting it into a vector (e.g., pBR322, pUC18 and pUC19) which functions in an E. coli strain.
  • Then, an E. coli strain (e.g., E. coli strain DH1, E. coli strain JM109 and E. coli strain HB101) which is transformed with the expression vector described above may be cultured in an appropriate medium to obtain the desired peptide. When a signal peptide of bacteria (e.g., signal peptide of pel B) is utilized, the desired peptide may be released in periplasm. Furthermore, a fusion protein with other peptide may be produced easily.
  • In a mammalian cell expression system, an expression vector is prepared by inserting a DNA, for example, having the nucleotide sequence shown in SEQ ID NO: 28, into the downstream of a proper promoter (e.g., SV40 promoter, LTR promoter and metallothionein promoter) in a proper vector (e.g., retro virus vector, papilloma virus vector, vaccinia virus vector and SV40 vector). Next, suitable mammalian cells (e.g., monkey COS-1 cells, COS-7 cells, Chinese hamster CHO cells, mouse L cells, 293 cells etc.) are transfected with the expression vector thus obtained, and then the transfected cells are cultured in an appropriate medium, the aimed peptide can be secreted into the culture medium.
  • A transformation of E. coli can be carried out in accordance with the method, for example, described in Proc. Natl. Acad. Sci. (USA) 69:2110, 1972 and Gene 17: 107, 1982.
  • A transfection of mammalian cells can be carried out in accordance with the method, for example, described in Saiboukougaku supl. 8:263 (New experimental protocol for cell technology), Shujun-sha, 1995 and Virology 52:456, 1973.
  • It is known that a bispecific antibody can be prepared directly by using recombinant DNA technology. For example, Alt et al. (FEBS Letter 454:90, 1999) reported the generation of a bispecific antibody (it is referred to a single-chain diabody) directed against carcinoembryonic antigen and E. coli beta-galactosidase. In said fragment, a heavy chain variable domain (VH) of one antibody is connected to a light chain variable domain (VL) of the other antibody with a short linker, which prevent the pairing of these two continuous domains on the same chain. Therefore, the VH and VL domains of this fragment are obliged to pair with each complementary VL and VH domain on other chain, and thereby form two antigen binding sites.
  • It is preferred that a peptide linker contains 3 to 12 amino acid residues, but it is not limited particularly to its amino acid sequence (Hudson et al., J. Immunol. Met. 231:177, 1999).
  • (3) The peptide thus obtained can be purified by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography.
  • Since bispecific antibody of the present invention also recognizes PD-1 specifically, it can be used for the purification and concentration of PD-1, for example, for affinity chromatography etc.
  • INDUSTRIAL APPLICABILITY Application for Pharmaceuticals
  • The greatest and important usage of a substance that specifically recognizes PD-1 by the present invention is for the medical treatment of the following disease.
  • A substance that specifically recognizes PD-1 by the present invention is useful for the medical treatment and/or prevention of diseases such as neurodegenerative disease (Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease etc.).
  • A substance that specifically recognizes PD-1 by the present invention is also useful for the medical treatment and/or prevention of diseases, in which PD-1 is involved and immune responses are enhanced, such as autoimmune diseases (glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis etc.), organ transplant rejection, and allergy.
  • A substance that specifically recognizes PD-1 by the present invention is also useful for the medical treatment and/or prevention of diseases, in which PD-1 is involved and immune responses are reduced, such as neoplasm and infections.
  • For the above mentioned usage, administration of the substance that specifically recognizes PD-1 by the present invention can be carried out in systemic or local, generally peroral or parenteral ways.
  • The dosage to be administered depends upon age, body weight, symptom, desired therapeutic effect, route of administration, and duration of the treatment etc. In human adults, one dose per person is generally between 0.1 mg and 100 mg by oral administration up to several times per day, or between 0.01 mg and 30 mg by parenteral administration (preferably intravenous administration) up to several times per day, or continuous administration between 1 and 24 hrs. per day into vein.
  • As mentioned above, the doses to be used depend upon various conditions. Therefore, there are cases in which doses lower than or greater than the ranges specified above may be used.
  • The compounds of the present invention may be administered as inner solid compositions or inner liquid compositions for oral administration, or as injections, liniments or suppositories etc. for parenteral administration.
  • Examples of Inner solid compositions for oral administration include compressed tablets, pills, capsules, dispersible powders and granules etc. Examples of capsules include hard capsules and soft capsules.
  • In such inner solid compositions, one or more of the active compound(s) remains intact, or is/are admixed with excipients (lactose, mannitol, glucose, microcrystalline cellulose and starch etc.), connecting agents (hydroxypropyl cellulose, polyvinylpyrrolidone, magnesium metasilicate aluminate, etc.), disintegrating agents (cellulose calcium glycolate etc.), lubricating agents (magnesium stearate etc.), stabilizing agents, assisting agents for dissolving (glutamic acid, asparaginic acid etc.) etc. to prepare pharmaceuticals by known methods. The pharmaceuticals may, if desired, be coated with coating agent (sugar, gelatin, hydroxypropyl cellulose or hydroxypropylmethyl cellulose phthalate etc.), or be coated with two or more films. Further, coating may include capsules of absorbable materials such as gelatin.
  • Inner liquid compositions for oral administration may contain pharmaceutically acceptable water-agents, suspensions, emulsions, syrups and elixirs etc. In such liquid compositions, one or more of the active compound(s) is/are resolved, suspended or emulsified in inert diluent (s) commonly used in the art (purified water, ethanol or mixture thereof etc.). Such liquid compositions may also comprise wetting agents, suspending agents, emulsifying agent, sweetening agents, flavoring agents, perfuming agents, preserving agents and buffer agents etc.
  • Injections for parenteral administration include solutions, suspensions and emulsions and solid injections, which are dissolved or suspended in solvent when it is used. In such compositions, one or more active compound(s) is/are dissolved, suspended or emulsified in a solvent. Solvents include distilled water for injection, physiological salt solution, plant oil, propylene glycol, polyethylene glycol, alcohol such as ethanol, and mixture thereof etc. Such compositions may comprise additional stabilizing agents, assisting agents for dissolving (glutamic acid, asparaginic acid, POLYSOLBATE80 (registered trademark) etc.), suspending agents, emulsifying agents, soothing agent, buffer agents, preserving agents etc. They may be manufactured or prepared by sterilization or by aseptic manipulation in a final process. They may also be manufactured in the form of sterile solid compositions such as freeze-dried compositions, and can be dissolved in sterile water or some other sterile solvent for injection immediately before use.
  • Other compositions for parenteral administration include liquids for external use, ointments, endermic liniments, aerosols, spray compositions, suppositories and pessaries for vaginal administration etc., which comprise one or more of the active compound(s) and may be prepared by known methods.
  • Spray compositions may comprise additional substances other than inert diluents generally used: e.g. stabilizing agents such as sodium hydrogen sulfate, buffer agents to give isotonicity, isotonic buffer such as sodium chloride, sodium citrate and citric acid. For preparation of such spray compositions, for example, the methods described in the U.S. Pat. No. 2,868,691 and U.S. Pat. No. 3,095,355 may be used.
  • Since PD-1 is involved in immune responses, the substance that specifically recognizes PD-1 by the present invention can also be used for the screening of substances, which are involved in immune responses, by measuring the expression of PD-1.
  • EFFECT OF THE INVENTION
  • A substance that specifically recognizes PD-1 by the present invention comprises a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker, is a superior substance recognizable both PD-1 and the membrane protein specifically and transmittable the signal of PD-1.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the results of FACS analysis using anti-PD-1/anti-TCR hybrid Fab antibodies;
  • FIG. 2 shows the effect of anti-PD-1/anti-TCR hybrid Fab antibodies on activated T cells;
  • FIG. 3 shows the inhibitory effect of anti-PD-1/anti-BCR hybrid Fab antibodies on the production of IL-2 by anti-BCR antibody stimulation;
  • FIG. 4 shows the effect of anti-PD-1/anti-BCR hybrid Fab antibodies on SHP-2 recruitment after anti-BCR antibody stimulation;
  • FIG. 5 shows the plasmid J43-2C11scDb-pSec/hygro B for the expression of J43-2C11 bispecific antibody; and
  • FIG. 6 shows the effect of J43-2C11 bispecific antibodies on the production of IFN-r by activated mouse spleen T cells.
  • BEST MODE FOR CARRYING OUT THE INVENTION
  • The present invention is more specifically explained by means of the following examples, but is not limited only to these examples.
  • Hereinafter, a substance in which anti-PD-1 antibody and anti-T cell receptor antibody are linked by a linker simply referred to as “anti-PD-1/anti-TCR hybrid antibody”, and a substance in which anti-PD-1 antibody and anti-B cell receptor antibody are linked by a linker “anti-PD-1/anti-BCR hybrid antibody”, respectively. A substance in which the FabSH portion of anti-PD-1 antibody and the FabSH portion of anti-T cell receptor antibody are linked by a linker hereinafter simply referred to as “anti-PD-1/anti-TCR hybrid Fab antibody”, and a substance in which the FabSH portion of anti-PD-1 antibody and the FabSH portion of anti-B cell receptor antibody are linked by a linker “anti-PD-1/anti-BCR hybrid Fab antibody”, respectively.
  • Example 1 (1) Preparation of Anti-PD-1/Anti-TCR Hybrid Antibodies (1-A) Introduction of Maleimide to Anti-Mouse CD3ε Monoclonal Antibodies
  • Anti-mouse CD3ε monoclonal antibodies were substituted with Sodium phosphate (0.1M, pH7.0) and Nacl (50 mM), then 200 times quantity of sulfo-EMCS (manufactured by Doj in Chemical) were added and incubated at 20° C. for 1 hour. Then the reaction mixture was size-fractionated by gel filtration using Sephacril S-300 [Sodium phosphate (0.1M, pH7.0)] and major peak fractions were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (1-B) Reduction of Anti-PD-1 Antibodies
  • Antibodies against mouse PD-1 (produced by the hybridoma cells named (J43) and deposited as international deposition Accession number FERM BP-8118) were substituted with Sodium phosphate (0.1M, pH6.0), added with 2-mercaptoethylamine (final concentration of 10 mM) and EDTA (final concentration of 1 mM), and reduced at 37° C. for 90 minutes. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-300 [Sodium phosphate (0.1M, pH6.0)] and major peak fractions (single chain fractions) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (1-C) Bridging of Maleimide-Conjugated Anti-Mouse CD3ε Monoclonal Antibodies and Reduced Anti-PD-1 Antibodies
  • Maleimide-conjugated anti-mouse CD3ε monoclonal antibodies and Reduced anti-PD-1 antibodies were mixed at the rate of 1:4 and incubated at 15° C. for 18 hours. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-300 [Sodium phosphate (0.1M, pH7.0)] and major peak fractions were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (2) Preparation of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies (2-A) Preparation of F(ab′)2 Fraction of Anti-PD-1 Antibodies
  • Antibodies against mouse PD-1 (produced by the hybridoma cells named (J43) and deposited as international deposition Accession number FERM BP-8118) were substituted with pepsin-buffer [(sodium acetate 0.1M, pH4.5), NaCl (0.1M)], added with pepsin (final concentration of 0.2 mg/ml), and digested for 48 hours at 37° C. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (2-B) Preparation of FabSH Fraction of Anti-PD-1 Antibodies
  • 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (2-C) Preparation of F(ab′)2 Fraction of Anti-Mouse CD3ε Monoclonal Antibodies
  • Anti-mouse CD3ε monoclonal antibodies (manufactured by Pharmingen) were substituted with pepsin-buffer [(sodium acetate 0.1M, pH4.5), NaCl (0.1M)], added with pepsin (final concentration of 0.2 mg/ml), and digested for 48 hours at 37° C. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (2-D) Preparation of FabSH Fraction of Anti-Mouse CD3ε Monoclonal Antibodies
  • 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (2-E) Bridging of FabSH Fraction of Anti-PD-1 Antibodies and FabSH Fraction of Anti-Mouse CD3ε Monoclonal Antibodies
  • Phenylenedimaleimide (manufactured by Aldrich) (final concentration of 4 mM) was added to the FabSH fraction of anti-PD-1 antibodies prepared in step (2-A), and incubated for 30 minutes at room temperature to prepare J43 Fabmal fraction. The J43 Fabmal fraction and the FabSH fraction of anti-mouse CD3ε monoclonal antibodies were mixed at the rate of 1:1.3 and incubated for 4 hours at room temperature. Next, an appropriate amount of Tris-HCl (1M, pH8.0) were added to make the pH of the reaction mixture 8.0, 2-mercaptoethanol (final concentration of 20 mM) was added and incubated at 30° C. for 30 minutes. Then iodoacetoamide (manufactured by Sigma) was added (final concentration of 25 mM) and incubated for additional 10 minutes at room temperature under light shielding.
  • Finally, the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (BsAb fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • Example 2 Confirmation of Reactivity of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies on Cell Surface Antigen (PD-1 and CD3)
  • 1×106 cells were recovered from mPD-1/A20IIA1.6 cells as a PD-1 positive and CD3 negative cell, and naive T cells prepared from mouse spleen cells as a PD-1 negative and CD3 positive cell, respectively. The cells were added with 91 μl of FACS buffer (PBS(−) containing 0.5% BSA, EDTA (2 mM) and 0.01% NaN3), 5 μl of mouse serum and 4 μl of hybrid antibodies (2 μg) and incubated for 30 minutes on ice. After washing with PBS(−) once, the cells were added with each 2 μl (1 μg) of second antibodies, fill upped to final 100 μl with FACS buffer, and incubated for 30 minutes on ice. Then they were analyzed by using FACScan. The results were shown in FIG. 1 (in the following figures, hybrid Fab antibody simply referred to as “HFAb”).
  • Analysis whether the prepared anti-PD-1/anti-TCR hybrid Fab antibodies actually react with the cell surface antigens by using FACSsort (manufactured by Becton Dickinson) showed the results that the antibodies reacted with both surface antigens.
  • Example 3 Effect of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies on Activated T Cells (A) Preparation of Spleen T Cells
  • Spleen was excised from BALB/c mouse. After red blood cells were hemolyzed, the spleen cells were washed once with PBS (−) and suspended in medium RPMI 1640 (10% FCS, antibiotics) (1×108 cells/ml). Next, T cells were prepared by using nylon fiber column (manufactured by WAKO) for T cell separation equilibrated with the medium.
  • (B) Effect of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies on Activated Spleen T Cells
  • Ninety six-well plates were coated with 0.5 μg/ml and 5 μg/ml of anti-CD3 antibodies (clone KT3, manufactured by Immuntech) at 37° C. for 3 hours. T cells (2×105 cells/well/200 μl) were seeded on the plates, anti-PD-1/anti-TCR hybrid antibodies (0.03, 0.1, 0.3, 1, 3 μg/100 ml) were added, and incubated in a CO2 incubator (at 37° C.). After 72 hours, cytokine (IFN-r, IL-2, IL-4 and IL-10) concentrations in the recovered culture supernatants were measured by using assay kit (manufactured by R & D System).
  • As the results shown in FIG. 2, after 72 hours when 3 μg of anti-PD-1/anti-TCR hybrid Fab antibodies were used, the results were obtained that the stimulation by the antibodies both in 0.5 μg/ml and 5 μg/ml suppressed the production of IFN-r, IL-4 and IL-10 dominantly.
  • Example 4 Preparation of Anti-PD-1/Anti-BCR Hybrid Fab Antibodies (A) Preparation of F(ab′)2 Fraction of Anti-PD-1 Antibodies
  • Antibodies against mouse PD-1 (the same antibodies as used in Example 1) were substituted with pepsin-buffer [(sodium acetate 0.1M, pH4.5), NaCl (0.1M)], added with pepsin (manufactured by SIGMA) (final concentration of 0.2 mg/ml), and digested at 37° C. for 48 hours. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 (manufactured by AmarshamParmacia) [Tris-HCl (0.2M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (B) Preparation of FabSH Fraction of Anti-PD-1 Antibodies
  • 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (C) Preparation of F(ab′)2 Fraction of Anti-IgG (H+L) Polyclonal Antibodies
  • Rabbit anti-mouse IgG (H+L) polyclonal antibodies (manufactured by Zymed) were substituted with pepsin-buffer [(sodium acetate 0.1M, pH4.5), NaCl (0.1M)], added with pepsin (final concentration of 0.2 mg/ml), and digested at 37° C. for 48 hours. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (D) Preparation of FabSH Fraction of Anti-IgG (H+L) Polyclonal Antibodies
  • 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • (E) Bridging of FabSH Fraction of Anti-PD-1 Antibodies and FabSH Fraction of Anti-IgG (H+L) Polyclonal Antibodies
  • Phenylenedimaleimide (manufactured by Aldrich) (final concentration of 4 mM) was added to the FabSH fraction of J43 anti-PD-1 antibodies, and incubated for 30 minutes at room temperature to prepare J43 Fabmal fraction. The J43 Fabmal fraction and the FabSH fraction of anti-IgG (H+L) polyclonal antibodies were mixed at the rate of 1:1.3 and incubated for 4 hours at room temperature. Next, an appropriate amount of Tris-HCl (1M, pH8.0) were added to make the pH of the reaction mixture 8.0, 2-mercaptoethanol (final concentration of 20 mM) was added and incubated at 30° C. for 30 minutes. Then iodoacetoamide (manufactured by SIGMA) was added (final concentration of 25 mM) and incubated for additional 10 minutes at room temperature under light shielding. Finally, the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (BsAb fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.
  • Example 5 Effect of Anti-PD-1/Anti-BCR Hybrid Fab Antibodies on B Cell Line
  • (A) Generation of A20IIA1.6 (B Cell Line) that was Forced to Express Mouse PD-1
  • (1) Construction of Expression Plasmid for Mouse PD-1
  • DNA fragments of mPD1-flag digested with EcoRI were inserted into the EcoRI site of a commercially available expression vector to construct the expression plasmid mPD1-pA.
  • (2) Transfection
  • A20IIA1.6 cells (1×107) in 325 μl of ice-cold RPMI1640 medium containing 15% FCS and the PD-1 expression plasmid linearized with ScaI in 10 μl of distilled water were incorporated into a Cuvette for electroporation (Gene Pulser Cuvette 0.4 cm electrode gap, 50, BIO RAD), and pulsed under the condition of 250V/960 μF (Gene Pulser, BIO RAD). After leaving at rest for 10 minutes at room temperature, the cells were suspended in 30 ml of medium (RPMI1640 containing 10% FBS, 50 μl of 2-mercaptoethanol, penicillin and streptomycin), diluted thirty fold further, and dispensed onto 96-well plates (103/100 μl/well). After 48 hours, the selection was initiated using final 3 μM of Puromycin to establish the cell line expressing mouse PD-1.
  • (B) Effect of Anti-PD-1/Anti-BCR Hybrid Fab Antibodies on A20IIA1.6 Cells that was Forced to Express Mouse PD-1
  • A20IIA1.6 cells that was forced to express mouse PD-1 were seeded on 96-well plates (5×105 cells/100 μl). Anti-PD-1/anti-BCR hybrid Fab antibodies (0, 1, 3, 10 μg/100 μl) were added, 10 minutes later 100 μl of anti-mouse IgG (H+L) F(ab′)2 (manufactured by Zymed) (final concentration of 0.3, 1, 3 μg/ml) were dispensed and cultivated for 12 hours in a CO2 incubator (at 37° C.). The culture supernatants were recovered and the concentrations of IL-2 in the culture supernatants were measured by using mouse IL-2 assay kit (manufactured by R & G System). The results were shown in FIG. 3.
  • The studies using various doses of anti-PD-1/anti-BCR hybrid Fab antibodies and anti-BCR antibodies F(ab′)2 showed the suppressive effects of the hybrid Fab antibodies in all cases, regardless of the concentration of anti-BCR antibodies F(ab′)2.
  • (C) Confirmation of SHP-2 Recruitment
  • Anti-PD-1/anti-BCR hybrid Fab antibodies (0, 1, 3, 10 μg/100 μl) were added to 3×106 of A20IIA1.6 cells (B cell line) that was forced to express mouse PD-1. After 10 minutes, 100 μl of anti-mouse IgG (H+L) F(ab′)2 were added and incubated for 5 minutes at room temperature. After discarding the supernatants by centrifugation, the cells were suspended in 200 μl of lysis buffer (composition: Tris-HCl (20 mM, pH7.4), NaCl (150 mM), Na2EDTA (1 mM), EGTA (1 mM), 1% Triton-X100, sodium pyrophosphate (2.5 mM), 1-sodium glycerophosphate (1 mM), Na3VO4 (1 mM), leupeptin (1 μg/ml) and PMSF (1 mM) and left at rest on ice. After 30 minutes, the supernatants were recovered by centrifugation, 20 μl of protein G-cepharose beads (manufactured by Amersham Bioscience) added, and incubated at 4° C. for 30 minutes. After recovering the supernatants by centrifugation, 20 μl of protein G-cepharose beads bound beforehand with anti-FLAG antibodies (manufactured by SIGMA) were added and mixed over night at 4° C.
  • The beads were washed five times with 400 μl of lysis buffer, added with 20 μl of lysis buffer and 20 μl of 2×SDS sample buffer, and boiled at 100° C. for 5 minutes. After discarding the beads by centrifugation, 15 μl of supernatants were subjected to 4-20% SDS-PAGE. After electrophoresis the gels were substituted with blotting buffer and transferred onto PVDF membrane (manufactured by BIO RAD). Then the membrane was blocked with Block Ace (manufactured by Dainippon Pharmaceuticals) for 1 hour at room temperature.
  • The membrane was incubated with anti-SHP-2 antibody (manufactured by SANTA CRUZ) diluted 1/200 for 1 hour at room temperature, then washed three times with TBS-T for 10 minutes. Next, the membrane was incubated with HRP-conjugated anti-rabbit Ig antibody (manufactured by Amersham Bioscience) diluted 1/2000 for 1 hour at room temperature, then washed three times with TBS-T for 10 minutes. Finally the membrane was emitted light by using ECT plus detection kit (manufactured by Amersham Bioscience) and analyzed using luminoimager LAS1000 plus (manufactured by FUJI Film).
  • Since anti-PD-1/anti-BCR hybrid Fab antibodies suppressed the production of IL-2 from anti-BCR antibody stimulated B cells, the evaluation whether the effects were caused by the recruitment of phosphatase SHP-2 to ITIM of PD-1 was carried out. As shown in FIG. 4, by adjusting the amount of samples with that of PD-1 as a control, the determination of the quantity of SHP-2 recruitment resulted in the obvious recruitment of SHP-2, in 1 and 10 μg, as compared with control hybrid antibodies.
  • Example 6 cDNA Cloning of Anti-Mouse PD-1 Antibody J43 (1) Preparation of Anti-Mouse PD-1 Antibody J43
  • Anti-mouse PD-1 antibody producing hybridoma (J43) cells were cultured in Hybridoma SFM medium (manufactured by Invitrogen) at 37° C. under 5% CO2, a few days after the culture supernatants of hybridoma cells were recovered. IgG fraction was purified from the culture supernatants recovered by using HiTrap Protein G (manufactured by Amersham Bioscience).
  • (2) Peptide Sequencing
  • J43 IgG was subjected to 10-20% SDS-PAGE. After electrophoresis the IgG was electrically transferred from the gel onto PVDF membrane (manufactured by BIO RAD). The membrane transferred was stained with coomassie, the membrane fraction containing the light chain of J43 IgG was removed, and amino terminal 15 residues of the light chain were determined by using peptide sequencer Procise492 (manufactured by Applied Biosystems) (SEQ ID NO:1).
  • (3) Extraction of RNA
  • 5×106 hybridoma cells were lysed with 1 ml of TRIzol (manufactured by Invitrogen). Total RNA was prepared according to the direction of attached document. mRNA was purified from total RNA thus prepared by using Oligotex-MAG mRNA Purification Kit (manufactured by Takara Shuzo).
  • (4) Cloning of Light Chain cDNA (3′RACE)
  • Degenerated primer (primer No. 1) was designed based on the amino terminal sequence (YELTQPPSASVNVGE) of the light chain determined by peptide sequencing. 3′RACE was carried out by using 3′-Full RACE Core Set (manufactured by Takara Shuzo) under the following conditions.
  • Primer No.1
    (SEQ ID NO: 2)
    5′-ta(c/t) ga(a/g) ct(g/a/t/c) ac(g/a/t/c) ca(a/g)
    cc(g/a/t/c) cc-3′
  • 1) Synthesis of first strand cDNA
    10 × RNA polymerase chain reaction (PCR) buffer 2
    J43 mRNA (50 ng/μl) 2
    Magnesium chloride (25 mM) 4
    dNTP mixture (each 10 mM) 2
    AMV Reverse Transcriptase XL (5 U/μl) 1
    Oligo dT-3sites Adapter primer (2.5 pmol/μl) 1
    Ribonuclease (RNase) inhibitor (40 U/μl) 0.5
    dH2O 7.5
    15 μl
    30° C. 10 min. → 50° C. 30 min. → 95° C. 5 min. → 4° C. 5 min.
  • 2) Polymerase chain reaction (PCR)
    First strand cDNA 1
    Primer No. 1 (20 pmol/μl) 1
    Anchorprimer 1
    dH2O 22
    One shot LA PCR Mix ™ 25
    50 μl
    95° C. 5 min. → (94° C. 29 sec., 50° C. 20 sec., 72° C. 60 sec.) × 30 cycles
  • The PCR products were subjected to electrophoresis using 1% Agarose gel, then the gel was stained by EtBr. DNA fragment was recovered from the gel by using MinElute Gel Extraction Kit (manufactured by Quiagen), the DNA fragment recovered was ligated to pGEM-T Easy Vector (manufactured by Promega) by using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo). E. coli DH5α was transformed with the ligated plasmid. Finally, the plasmid was purified from E. coli and J43 IgG light chain cDNA was sequenced (SEQ ID NO:3). The deduced amino acid sequence of the cDNA is shown in sequence listing (SEQ ID NO:4)
  • (5) Cloning of Heavy Chain cDNA (5′RACE)
  • To perform 5′RACE, primers for constant region were designed based upon the reported information of hamster IgG heavy chain cDNA sequence (GenBank Accession No. U17166). 5′RACE was carried out by using 5′-Full RACE Core Set (manufactured by Takara Shuzo) under the following conditions.
  • Primer No.2
    5′-ccc aag agg tca gga gtt gga-3′ (SEQ ID NO: 5)
    (5′ phosphorylated)
    Primer No.3
    5′-ttg acc agg cat ccc agg gtc-3′ (SEQ ID NO: 6)
    Primer No.4
    5′-cgt aag ctg gaa ctc tgg agc-3′ (SEQ ID NO: 7)
    Primer No.5
    5′-tgg ttg tgc tgt cac agg cag-3′ (SEQ ID NO: 8)
    Primer No.6
    5′-tgc aca cct tcc cat ctg tcc t-3′ (SEQ ID NO: 9)
  • 1) Synthesis of first strand cDNA
    J43 total RNA (2 μg/μl) 2
    10 × RNA polymerase chain reaction (PCR) buffer 1.5
    Ribonuclease (RNase) inhibitor (40 U/μl) 0.5
    AMV Reverse Transcriptase XL (5 U/μl) 1
    Primer No. 2 (100 pmol/μl) 2
    dH2O 8
    15 μl
    30° C. 10 min. → 50° C. 40 min. → 30° C. 2 min.
  • 2) Degeneration of hybrid RNA
    First strand cDNA 15
    5 × Hybrid RNA Degeneration buffer 15
    dH2O 45
    Ribonuclease H (RNaseH) 1
    76 μl
    30° C. 1 hour
  • After the reaction, ethanol precipitation was carried out.
  • 3) Cyclization of single chain cDNA by ligation
    5 × RNA (ssDNA) Ligation buffer 8
    40% Polyethylene glycol (PEG) #6000 20
    dH2O 12
    Pellet after ethanol precipitation
    T4 RNA ligase 1
    41 μl
    15° C. 15 hours
  • 4) First PCR
    Sample after ligation 1
    Primer No. 3 (5 pmol/μl) 2
    Primer No. 4 (5 pmol/μl) 2
    dH2O 20
    One shot LA PCR Mix ™ 25
    50 μl
    94° C. 3 min. → (94° C. 30 sec., 52° C. 30 sec., 72° C. 120 sec.) × 25 cycles
  • 5) Second PCR
    Product of first PCR 2
    Primer No. 5 (5 pmol/μl) 2
    Primer No. 6 (5 pmol/μl) 2
    dH2O 20
    One shot LA PCR Mix ™ 25
    50 μl
    94° C. 3 min. → (94° C. 30 sec., 52° C. 30 sec., 72° C. 120 sec.) × 30 cycles
  • The PCR products were subjected to electrophoresis using 1% Agarose gel, then the gel was stained by EtBr. DNA fragment was recovered from the gel by using MinElute Gel Extraction Kit (manufactured by Quiagen), the DNA fragment recovered was ligated to pGEM-T Easy Vector (manufactured by Promega) by using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo). E. coli DH5α was transformed with the ligated plasmid. Finally, the plasmid was purified from E. coli and J43 IgG heavy chain cDNA was sequenced (SEQ ID NO:10). The deduced amino acid sequence of the cDNA is shown in sequence listing (SEQ ID NO:11)
  • Example 7 cDNA Cloning of Anti-Mouse CD3ε Antibody (1) Preparation of RNA
  • Anti-mouse CD3ε antibody producing hybridoma (145-2C11: manufactured by Pharmingen) cells were cultured in Hybridoma SFM medium (manufactured by Invitrogen) at 37° C. under 5% CO2, a few days after 5×106 hybrisoma cells were lysed with 1 ml of TRIzol (manufactured by Invitrogen). Total RNA was prepared according to the direction of attached document.
  • (1) Preparation of cDNA Library
  • cDNA was synthesized from 2.5 μg of total RNA extracted from hybridoma (145-2C11) cells by oligo-dT prime method using Ready-To-Go You-Prime First-Strand Beads (manufactured by Amersham Pharmacia). Operations and procedures were followed by the instructions of attached document.
  • (3) cDNA Cloning of Heavy and Light Chains
  • Based upon the reported cDNA sequence information of heavy chain variable region of hybridoma 145-2C11 (GenBank Accession No. AF000357), primers No. 7 and No. 8 were designed. Also, based upon the cDNA sequence information of light chain variable region of 145-2C11 (GenBank Accession No. AF000356), primers No. 9 and No. 10 were designed. PCR was carried out using these primers and the cDNA library from hybridoma 145-2C11 as a template.
  • Primer No.7
    (SEQ ID NO: 12)
    5′-gag gtg cag ctg gtg gag tct-3′
    Primer No.8
    (SEQ ID NO: 13)
    5′-tga gga gac ggt gac cat ggt t-3′
    Primer No.9
    (SEQ ID NO: 14)
    5′-gac atc cag atg acc cag tct c-3′
    Primer No.10
    (SEQ ID NO: 15)
    5′-ttt gat ttc cag ctt ggt gcc ag-3′
  • cDNA library 2
    Primer No. 7 or No. 9 (5 pmol/μl) 2
    Primer No. 8 or No. 10 (5 pmol/μl) 2
    dH2O 20
    One shot LA PCR Mix ™ 25
    50 μl
    (94° C. 30 sec., 52° C. 30 sec., 72° C. 120 sec.) × 30 cycles
  • The PCR products were subjected to electrophoresis using 1% Agarose gel, then the gel was stained by EtBr. DNA fragments were recovered from the gel by using MinElute Gel Extraction Kit (manufactured by Quiagen), the DNA fragments recovered was ligated to pGEM-T Easy Vector (manufactured by Promega) by using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo). E. coli DH5α were transformed with the ligated plasmids. Finally, the plasmids were purified from E. coli, and both DNA were sequenced. It was confirmed that their sequences were identical to those of GenBank Accession No. AF00357 and GenBank Accession No. AF000356, respectively.
  • Example 8 Constraction of Expression Plasmid for J43-2C11 Bispecific Antibody
  • J43 IgG heavy chain cDNA and 145-2C11 IgG light chain cDNA were connected by PCR using linker No. 1, No. 2, primer No. 11 and No. 12 to prepare fragment 1 (see FIG. 5). Next, 145-2C11 IgG light chain cDNA and 145-2C11 IgG heavy chain cDNA were connected by PCR using linker No. 3, No. 4, primer No. 13 and No. 14 to prepare fragment 2 (see FIG. 5). 145-2C11 IgG heavy chain cDNA and J43 IgG light chain cDNA were connected by PCR using linker No. 5, No. 6, primer No. 15 and No. 16 to prepare fragment 3 (see FIG. 5).
  • Primer No.11
    (SEQ ID NO: 16)
    5′-ttt gaa ttc aga ggt gcg gct tct gga gtc t-3′
    Primer No.12
    (SEQ ID NO: 17)
    5′-gat cag gag ctt agg agc ttt cc-3′
    Primer No.13
    (SEQ ID NO: 18)
    5′-cag gcc agt cag gac att agc aa-3′
    Primer No.14
    (SEQ ID NO: 19)
    5′-taa tgt atg cga ccg act cca gc-3′
    Primer No.15
    (SEQ ID NO: 20)
    5′-tga ggc ctc tgg att cac ctt ca-3′
    Primer No.16
    (SEQ ID NO: 21)
    5′-aaa aaa aaa ctc gag gac cta gga cgg tga gct ggg
    t-3′
    Linker No.1
    (SEQ ID NO: 22)
    5′-agg gac cca agt cac tgt ctc ctc agg tgg agg cgg
    ttc aga cat cca gat gac cca gtc tcc at-3′
    Linker No.2
    (SEQ ID NO: 23)
    5′-tcc ctg ggt tca gtg aca gag gag tcc acc tcc gcc
    aag tct gta ggt cta ctg ggt cag agg ta-3′
    Linker No.3
    (SEQ ID NO: 24)
    5′-acc tgg cac caa gct gga aat caa agg tgg agg cgg
    ttc agg cgg agg tgg ctc tgg cgg tgg cgg atc gga
    ggt gca gct ggt gga gtc tgg gg-3′
    Linker No.4
    (SEQ ID NO: 25)
    5′-tgg acc gtg gtt cga cct tta gtt tcc acc tcc gcc
    aag tcc gcc tcc acc gag acc gcc acc gcc tag cct
    cca cgt cga cca cct cag acc cc-3′
    Linker No.5
    (SEQ ID NO: 26)
    5′-agg aac cat ggt cac cgt ctc ctc agg tgg agg cgg
    ttc ata tga gct gac tca gcc acc ttc ag-3′
    Linker No.6
    (SEQ ID NO: 27)
    5′-tcc ttg gta cca gtg gca gag gag tcc acc tcc gcc
    aag tat act cga ctg agt cgg tgg aag tc-3′
  • PCR condition for fragments 1, 2 and 3
    First PCR
    Template
    1 2
    Template 2 2
    Linker (100 ng/μl) 2
    Linker (100 ng/μl) 2
    dH2O 17
    One shot LA PCR Mix ™ 25
    50 μl
    95° C. 5 min. → (94° C. 30 sec., 40° C. 30 sec., 72° C. 60 sec.) × 20 cycles
  • Second PCR
    Product of first PCR 5
    Primer (5 pmol/μl) 2
    Primer (5 pmol/μl) 2
    dH2O 16
    One shot LA PCR Mix ™ 25
    50 μl
    95° C. 5 min. → (94° C. 30 sec., 50° C. 30 sec., 72° C. 60 sec.) × 30 cycles
  • The DNA fragments 1, 2 and 3 and plasmid pBluescriptII SK(+) (manufactured by StrateGene) were digested with restriction enzymes EcoRI/KpnI, KpnI/SphI, SphI/XhoI, EcoRI/XhoI, respectively. After electrophoresis on 1% agarose gel, the DNA fragments were purified from the gel using MinElute Gel Extraction Kit. Next, these three fragments and plasmid pBluescriptII SK(+) (manufactured by StrateGene) were connected using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo), then E. coli DH5 (were transformed with the connected plasmid. Plasmid J43-2C11scDb-pBluescriptII SK(+) was prepared from E. coli, then the nucleotide sequence of the insert was determined (SEQ ID NO:28). The deduced amino acid sequence is shown in sequence listing (SEQ ID NO:29).
  • J43-2CllscDb-pBluescriptII SK(+) was digested with restriction enzymes BamHI and XhoI, the BamHI-XhoI fragment out of BamHI-XhoI and BamHI-BamHI fragments generated by the digestion was connected with BamHI/XhoI digested mammalian expression vector pSecTag2/Hygro B (manufactured by Invitrogen). Next, the pSecTag2/Hygro B connected with BamHI-XhoI fragment was digested again with restriction enzyme BamHI, and connected with the other BamHI-BamHI fragment. E. coliDH5α were transformed with the connected plasmid. Finally, Plasmid J43-2C11scDb-pSec/Hygro B was prepared from E. coli, then the nucleotide sequence of thus prepared J43-2C11 bispecific antibody was determined (SEQ ID NO:30). The deduced amino acid sequence is shown in sequence listing (SEQ ID NO:31).
  • Example 9 Expression of J43-2C11 Bispecific Antibody
  • 6×106 293 T cells were suspended in 20 ml of medium (DMEM containing 10% FBS) and seeded into 150 mm dish coated with Type Ic collagen. Next day, the cells were washed with 10 ml of DMEM and transfected with J43-2C11scDb-pSec/Hygro B by using LipofectAMINE-plus. After 3 hours, 5 ml of DMEM containing 40% FBS were added to the cells. At day 2, the cells were washed with 10 ml of DMEM, added 20 ml of new DMEM, and the culture supernatant was recovered at day 4.
  • The cells were discarded by centrifugation, then the supernatant was filtrated using 0.22 μm PVDF filter. The supernatant was enclosed in dialysis tube, dialyzed against PBS containing 40% PEG20000, and concentrated. The concentrated supernatant was purified using HiTrap chelating HP column (manufactured by Amersham Farmacia). To purify further, the antibodies were purified by gel-filtration using Hiprep 16/60 Sephacryl S-200 High Resolution (manufactured by Amersham Farmacia).
  • Example 10 Suppression of T Cell Activation
  • Spleen was removed from BALB/c mouse, and cells were prepared using CellStrainer (70 μm Nyron). The cells were recovered by centrifugation, and red blood cells were hemolyzed by the addition of hemolysis buffer [NH4Cl (0.8%), KCO3 (0.1%) and EDTA (1 mM)]. The cells were washed with PBS (−) once, T cells were enriched by using mouse CD3+ T cell enrichment column kit (manufactured by R&D), and suspended in medium (RPMI1640 containing 10% FBS) in the proportion of 5×106 cells/ml. The T cells thus prepared were seeded on 96-well plates (2×105 cells/100 μl/well), which were coated preliminarily with 5 μg/ml of anti-CD3 antibodies (clone KT3) at 37° C. for 3 hours, added with J43-2C11 bispecific antibody diluted in the medium (0.01, 0.03, 0.1, 0.3, 1 and 3 μg/100 μl), and cultivated for 72 hours at 37° C. under 5% CO2. After 72 hours, the culture supernatants were recovered, and the concentrations of IFN-r in the supernatants were determined by using Quantikine Immunoassay Kit (manufactured by R&D).
  • As shown in FIG. 6, J43-2C11 bispecific antibody suppressed dose dependently the production of IFN-r from activated mouse spleen T cells in vitro.

Claims (6)

1. An isolated polypeptide consisting of the amino acid sequence of SEQ ID NO: 29.
2. An isolated polypeptide consisting of the amino acid sequence of SEQ ID NO: 31.
3. A cDNA consisting of the polynucleotide encoding the amino acid sequence of SEQ ID NO: 29.
4. A cDNA consisting of the polynucleotide encoding the amino acid sequence of SEQ ID NO: 31.
5. A cDNA consisting of the polynucleotide sequence of SEQ ID NO: 28.
6. A cDNA consisting of the polynucleotide sequence of SEQ ID NO: 28.
US12/057,637 2001-07-31 2008-03-28 Substance that specifically recognizes PD-1 Expired - Lifetime US7858746B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/057,637 US7858746B2 (en) 2001-07-31 2008-03-28 Substance that specifically recognizes PD-1

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JPP.2001-232303 2001-07-31
JP2001232303 2001-07-31
US10/485,466 US20040241745A1 (en) 2001-07-31 2002-07-30 Substance specific to pd-1
PCT/JP2002/007735 WO2003011911A1 (en) 2001-07-31 2002-07-30 Substance specific to pd-1
US12/057,637 US7858746B2 (en) 2001-07-31 2008-03-28 Substance that specifically recognizes PD-1

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US10/485,466 Division US20040241745A1 (en) 2001-07-31 2002-07-30 Substance specific to pd-1
PCT/JP2002/007735 Division WO2003011911A1 (en) 2001-07-31 2002-07-30 Substance specific to pd-1
US10485466 Division 2002-07-30

Publications (2)

Publication Number Publication Date
US20090076250A1 true US20090076250A1 (en) 2009-03-19
US7858746B2 US7858746B2 (en) 2010-12-28

Family

ID=19064242

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/485,466 Abandoned US20040241745A1 (en) 2001-07-31 2002-07-30 Substance specific to pd-1
US12/057,637 Expired - Lifetime US7858746B2 (en) 2001-07-31 2008-03-28 Substance that specifically recognizes PD-1

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/485,466 Abandoned US20040241745A1 (en) 2001-07-31 2002-07-30 Substance specific to pd-1

Country Status (5)

Country Link
US (2) US20040241745A1 (en)
EP (1) EP1445264B1 (en)
JP (1) JP4249013B2 (en)
AT (1) ATE524495T1 (en)
WO (1) WO2003011911A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100266617A1 (en) * 2007-06-18 2010-10-21 N.V. Organon Antibodies to human programmed death receptor pd-1
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2014194293A1 (en) 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
US9683048B2 (en) 2014-01-24 2017-06-20 Novartis Ag Antibody molecules to PD-1 and uses thereof
US9701749B2 (en) 2011-08-11 2017-07-11 Ono Pharmaceutical Co., Ltd. Therapeutic agent for autoimmune diseases comprising PD-1 agonist
CN107353326A (en) * 2017-05-09 2017-11-17 中山大学附属口腔医院 Non-antibody binding proteins and its application with reference to the acceptors of PD 1
US9914783B1 (en) 2016-09-14 2018-03-13 Abbvie Biotherapeutics Inc. Anti-PD-1 antibodies and their uses
CN109160949A (en) * 2018-07-23 2019-01-08 中国医学科学院血液病医院(血液学研究所) A kind of anti-human PD-1 monoclonal antibody of mouse and application
US10443061B2 (en) * 2014-05-01 2019-10-15 Anaeropharma Science, Inc. Heterologous polypeptide expression cassette
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US11091550B2 (en) 2018-02-09 2021-08-17 Ono Pharmaceutical Co., Ltd. Bispecific antibody
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
WO2022213414A1 (en) * 2021-04-09 2022-10-13 中山大学附属口腔医院 Receptor pd-1/ligand pd-l1 dual-targeting non-antibody binding polypeptide or derivative thereof, and application thereof

Families Citing this family (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2206517T3 (en) 2002-07-03 2023-11-06 Ono Pharmaceutical Co Immunopotentiating compositions comprising anti-PD-L1 antibodies
WO2004056875A1 (en) * 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2004072286A1 (en) * 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
CA2607147C (en) * 2005-05-09 2018-07-17 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1 (pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
JP5252635B2 (en) 2005-07-01 2013-07-31 メダレックス インコーポレーティッド Human monoclonal antibody against programmed death ligand 1 (PD-L1)
WO2007122815A1 (en) * 2006-04-14 2007-11-01 Ono Pharmaceutical Co., Ltd. Bivalent antibody directed against bir1
KR20110014607A (en) 2008-04-29 2011-02-11 아보트 러보러터리즈 Dual variable domain immunoglobulins and uses thereof
TW201008580A (en) 2008-06-03 2010-03-01 Abbott Lab Dual variable domain immunoglobulin and uses thereof
BRPI0913366A8 (en) 2008-06-03 2017-07-11 Abbott Lab DUAL VARIABLE DOMAIN IMMUNOGLOBULINS AND THEIR USES
US8822645B2 (en) 2008-07-08 2014-09-02 Abbvie Inc. Prostaglandin E2 dual variable domain immunoglobulins and uses thereof
LT4209510T (en) 2008-12-09 2024-03-12 F. Hoffmann-La Roche Ag Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011028811A2 (en) * 2009-09-01 2011-03-10 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
CN102666875A (en) * 2009-10-15 2012-09-12 雅培制药有限公司 Dual variable domain immunoglobulins and uses thereof
UY32979A (en) 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
CN103298834A (en) 2010-08-03 2013-09-11 Abbvie公司 Dual variable domain immunoglobulins and uses thereof
US9046513B2 (en) 2010-08-26 2015-06-02 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
JP2015508994A (en) 2011-12-30 2015-03-26 アッヴィ・インコーポレイテッド Dual variable domain immunoglobulins against IL-13 and / or IL-17
RU2636043C2 (en) 2012-11-01 2017-11-17 Эббви Инк. Anti-vegf/dll4-immunoglobulins with double variable domains and their application
HUE050787T2 (en) 2013-02-26 2021-01-28 Memorial Sloan Kettering Cancer Center Compositions and methods for immunotherapy
AU2014227732A1 (en) 2013-03-15 2015-09-17 Abbvie Inc. Dual specific binding proteins directed against IL-1 beta and IL-17
MX2015015037A (en) 2013-05-02 2016-07-08 Anaptysbio Inc Antibodies directed against programmed death-1 (pd-1).
CN105085680A (en) * 2014-05-23 2015-11-25 复旦大学 Humanized anti-PD-1 and c-MET bispecific antibody, and preparation method and application thereof
TWI693232B (en) 2014-06-26 2020-05-11 美商宏觀基因股份有限公司 Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
HUE045108T2 (en) * 2014-07-16 2019-12-30 Transgene Sa Oncolytic virus for expression of immune checkpoint modulators
US9982052B2 (en) 2014-08-05 2018-05-29 MabQuest, SA Immunological reagents
AU2015298356B2 (en) 2014-08-05 2020-11-19 MabQuest SA Immunological reagents binding to pd-1
CN115040532A (en) 2014-10-10 2022-09-13 伊黛拉制药有限公司 Treatment of cancer using TLR9 agonists and checkpoint inhibitors
WO2016094881A2 (en) 2014-12-11 2016-06-16 Abbvie Inc. Lrp-8 binding proteins
TWI773646B (en) 2015-06-08 2022-08-11 美商宏觀基因股份有限公司 Lag-3-binding molecules and methods of use thereof
TW201709929A (en) 2015-06-12 2017-03-16 宏觀基因股份有限公司 Combination therapy for the treatment of cancer
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
CN114591433A (en) 2015-07-13 2022-06-07 西托姆克斯治疗公司 anti-PD-1 antibodies, activatable anti-PD-1 antibodies, and methods of use thereof
BR112018000917A2 (en) 2015-07-16 2018-09-11 Bioxcel Therapeutics Inc innovative approach to cancer treatment through immunomodulation
GEP20227419B (en) 2015-07-30 2022-10-10 Macrogenics Inc Pd-1-binding molecules and methods of use thereof
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
KR20220131277A (en) 2015-09-01 2022-09-27 아게누스 인코포레이티드 Anti-pd-1 antibodies and methods of use thereof
KR20180054824A (en) 2015-09-29 2018-05-24 셀진 코포레이션 PD-1 binding protein and method of use thereof
AU2016334041B2 (en) 2015-10-08 2023-02-09 Macrogenics, Inc. Combination therapy for the treatment of cancer
WO2017106061A1 (en) 2015-12-14 2017-06-22 Macrogenics, Inc. Bispecific molecules having immunoreactivity with pd-1 and ctla-4, and methods of use thereof
EP3405497A2 (en) 2016-01-22 2018-11-28 Mabquest SA Immunological reagents
US11214617B2 (en) 2016-01-22 2022-01-04 MabQuest SA Immunological reagents
CA3014001A1 (en) 2016-02-25 2017-08-31 Cell Medica Switzerland Ag Binding members to pd-l1
CA3016474A1 (en) 2016-03-15 2017-09-21 Mersana Therapeutics, Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
CN109952317A (en) 2016-09-19 2019-06-28 细胞基因公司 Use the method for PD-1 binding protein treatment immune disorders
US10766958B2 (en) 2016-09-19 2020-09-08 Celgene Corporation Methods of treating vitiligo using PD-1 binding antibodies
SG11201903835WA (en) 2016-11-01 2019-05-30 Anaptysbio Inc Antibodies directed against programmed death- 1 (pd-1)
US11135307B2 (en) 2016-11-23 2021-10-05 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
JP7042753B2 (en) 2016-12-28 2022-03-28 学校法人近畿大学 How to select individuals to receive immune checkpoint inhibitors
WO2018129559A1 (en) 2017-01-09 2018-07-12 Tesaro, Inc. Methods of treating cancer with anti-pd-1 antibodies
WO2018160538A1 (en) 2017-02-28 2018-09-07 Mersana Therapeutics, Inc. Combination therapies of her2-targeted antibody-drug conjugates
SG11201907948TA (en) * 2017-03-29 2019-09-27 Celgene Corp Formulations comprising pd-1 binding proteins and methods of making thereof
WO2019070047A1 (en) 2017-10-06 2019-04-11 小野薬品工業株式会社 Bispecific antibody
WO2019104289A1 (en) 2017-11-27 2019-05-31 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
US20220305127A1 (en) 2017-12-21 2022-09-29 Mersana Therapeutics, Inc. Pyrrolobenzodiazepine antibody conjugates
EP3876986A4 (en) * 2018-09-18 2022-06-01 Pandion Operations, Inc. Targeted immunotolerance
BR112021008012A2 (en) 2018-10-29 2021-11-03 Mersana Therapeutics Inc Engineered cysteine antibody-drug conjugates with peptide-containing linkers
TW202102544A (en) 2019-04-04 2021-01-16 日商小野藥品工業股份有限公司 Bispecific antibody
JP6881658B2 (en) * 2019-07-05 2021-06-02 小野薬品工業株式会社 Blood cancer treatment with PD-1 / CD3 bispecific protein
KR20220039720A (en) 2019-07-30 2022-03-29 오노 야꾸힝 고교 가부시키가이샤 bispecific antibody
JPWO2021025140A1 (en) 2019-08-08 2021-02-11
IL286430A (en) * 2021-09-14 2023-04-01 Yeda Res & Dev Multispecific antibodies for use in treating diseases

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5629204A (en) * 1994-03-01 1997-05-13 Ono Pharmaceutical Co., Ltd. Peptide related to human programmed cell death and DNA encoding it
US7029674B2 (en) * 2001-04-02 2006-04-18 Wyeth Methods for downmodulating immune cells using an antibody to PD-1

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU228477B1 (en) * 1999-08-23 2013-03-28 Dana Farber Cancer Inst Inc Pd-1, a receptor for b7-4, and uses therefor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5629204A (en) * 1994-03-01 1997-05-13 Ono Pharmaceutical Co., Ltd. Peptide related to human programmed cell death and DNA encoding it
US7029674B2 (en) * 2001-04-02 2006-04-18 Wyeth Methods for downmodulating immune cells using an antibody to PD-1

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9834605B2 (en) 2007-06-18 2017-12-05 Merck Sharpe & Dohme B.V. Antibodies to human programmed death receptor PD-1
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
US8900587B2 (en) 2007-06-18 2014-12-02 Merck Sharp & Dohme Corp. Antibodies to human programmed death receptor PD-1
US8952136B2 (en) 2007-06-18 2015-02-10 Merck Sharp & Dohme B.V. Antibodies to human programmed death receptor PD-1
US20100266617A1 (en) * 2007-06-18 2010-10-21 N.V. Organon Antibodies to human programmed death receptor pd-1
US11117961B2 (en) 2007-06-18 2021-09-14 Merck Sharp & Dohme B.V. Antibodies to human programmed death receptor PD-1
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
US9205148B2 (en) 2011-04-20 2015-12-08 Medimmune, Llc Antibodies and other molecules that bind B7-H1 and PD-1
EP3403672A1 (en) 2011-04-20 2018-11-21 Medlmmune, LLC Antibodies and other molecules that bind b7-h1 and pd-1
US10647770B2 (en) 2011-08-11 2020-05-12 Ono Pharmaceutical Co., Ltd. Therapeutic agent for autoimmune diseases comprising PD-1 agonist
US9701749B2 (en) 2011-08-11 2017-07-11 Ono Pharmaceutical Co., Ltd. Therapeutic agent for autoimmune diseases comprising PD-1 agonist
WO2014194293A1 (en) 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
US11708412B2 (en) 2013-09-26 2023-07-25 Novartis Ag Methods for treating hematologic cancers
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US11827704B2 (en) 2014-01-24 2023-11-28 Novartis Ag Antibody molecules to PD-1 and uses thereof
US9815898B2 (en) 2014-01-24 2017-11-14 Novartis Ag Antibody molecules to PD-1 and uses thereof
US9683048B2 (en) 2014-01-24 2017-06-20 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10981990B2 (en) 2014-01-31 2021-04-20 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11155620B2 (en) 2014-01-31 2021-10-26 Novartis Ag Method of detecting TIM-3 using antibody molecules to TIM-3
US10443061B2 (en) * 2014-05-01 2019-10-15 Anaeropharma Science, Inc. Heterologous polypeptide expression cassette
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US10730953B2 (en) 2016-09-14 2020-08-04 Abbvie Biotherapeutics Inc. Anti-PD-1 antibodies and their uses
US9914783B1 (en) 2016-09-14 2018-03-13 Abbvie Biotherapeutics Inc. Anti-PD-1 antibodies and their uses
CN107353326A (en) * 2017-05-09 2017-11-17 中山大学附属口腔医院 Non-antibody binding proteins and its application with reference to the acceptors of PD 1
US11091550B2 (en) 2018-02-09 2021-08-17 Ono Pharmaceutical Co., Ltd. Bispecific antibody
CN109160949A (en) * 2018-07-23 2019-01-08 中国医学科学院血液病医院(血液学研究所) A kind of anti-human PD-1 monoclonal antibody of mouse and application
WO2022213414A1 (en) * 2021-04-09 2022-10-13 中山大学附属口腔医院 Receptor pd-1/ligand pd-l1 dual-targeting non-antibody binding polypeptide or derivative thereof, and application thereof

Also Published As

Publication number Publication date
EP1445264B1 (en) 2011-09-14
JPWO2003011911A1 (en) 2004-11-18
JP4249013B2 (en) 2009-04-02
EP1445264A1 (en) 2004-08-11
EP1445264A4 (en) 2006-01-18
US20040241745A1 (en) 2004-12-02
WO2003011911A1 (en) 2003-02-13
ATE524495T1 (en) 2011-09-15
US7858746B2 (en) 2010-12-28

Similar Documents

Publication Publication Date Title
US7858746B2 (en) Substance that specifically recognizes PD-1
US9783609B2 (en) Substance specific to human PD-1
US8329178B2 (en) Antibodies against CXCR4 and methods of use thereof
MX2011000501A (en) Compositions monovalent for cd28 binding and methods of use.
AU7048691A (en) Cd4 specific recombinant antibody
CN107847592B (en) Antibodies against human CSF-1R for inducing lymphocytosis in lymphomas or leukemias
AU2002226086B2 (en) Silensed anti-CD28 antibodies and use thereof
CN113603780B (en) Therapeutic agents and methods for enhancing immune responses in tumor microenvironments
AU2002226086A1 (en) Silensed anti-CD28 antibodies and use thereof
RU2736650C2 (en) Activator of regulatory t cells and use thereof
CA3157611A1 (en) Anti-pd-l1/anti-b7-h3 multispecific antibodies and uses thereof
US20230056815A1 (en) Antibodies to feline mcdonough sarcoma (fms)-like tyrosine kinase 3 receptor ligand (flt3l) and uses thereof for treating autoimmune and inflammatory diseases
US20040116675A1 (en) Silenced anti-cd28 antibodies and use thereof
KR20010074697A (en) Immunological reagent specifically interacting with the extracellular domain of the human zeta chain
Mohseni Nodehi Improved Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) of Affinity Maturated and Fc-Engineered Antibodies Directed Against the AML Stem Cell Antigen CD96

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552)

Year of fee payment: 8

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1553); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 12